67 results on '"Pegoraro, S."'
Search Results
2. The impact of the complex interaction of basic mechanisms on reading decline in healthy aging
- Author
-
Pegoraro, S, Facchin, A, Luchesa, F, Rolandi, E, Guaita, A, Arduino, L, Daini, R, Pegoraro, S, Facchin, A, Luchesa, F, Rolandi, E, Guaita, A, Arduino, L, and Daini, R
- Published
- 2024
3. Regression-based normative data for Corsi Span and Supraspan learning and recall among Italian adults
- Author
-
Facchin, A, Pegoraro, S, Rigoli, M, Rizzi, E, Strina, V, Barera, S, Castiglieri, G, Daini, R, Guarnerio, C, Facchin, A, Pegoraro, S, Rigoli, M, Rizzi, E, Strina, V, Barera, S, Castiglieri, G, Daini, R, and Guarnerio, C
- Abstract
Introduction: The Corsi Block Tapping Test, or Corsi Span (CS), is a widely used task to measure visuospatial short-term and working memory. The same setup can be used to administer the Corsi SupraSpan Learning (CSSL) and Recall (CSSR), tests assessing visuospatial long-term memory. While the CS has relatively recent normative data, those of the CSSL are outdated For CSSR, no normative data are available. Given this critical lack, our study aimed to provide updated norms for CS, CSSL, and specifically for the recall delayed phase (CSSR). Materials and methods: A sample of 340 healthy participants, aged between 20 and 89, took part in the study. Norms were developed using a regression approach and defined using rank equivalent scores and percentiles. Results: Age and education influenced Corsi's Span, while SupraSpan Learning and Recall were influenced by age, education, and span. The comparison with previous norms for Span and SupraSpan Learning shows a high level of agreement. Conclusions: This study provides integrated norms to evaluate visuospatial memory in all aspects of immediate recall, long-term learning and delayed recall. Its use is needed to assess specific neuropsychological deficits, dissociate visuospatial versus verbal memory deficits and allow the evaluation of memory in patients with limited verbal abilities.
- Published
- 2024
4. Clinical course of isolated distal deep vein thrombosis in patients with active cancer: a multicenter cohort study
- Author
-
Dentali, F., Pegoraro, S., Barco, S., di Minno, M.N.D., Mastroiacovo, D., Pomero, F., Lodigiani, C., Bagna, F., Sartori, M., Barillari, G., Mumoli, N., Napolitano, M., Passamonti, S.M., Benedetti, R., Ageno, W., and Di Nisio, M.
- Published
- 2017
- Full Text
- View/download PDF
5. Effetti di un nuovo training cognitivo multidominio con preattivazione frontale nella popolazione anziana con Mild Cognitive Impairment (MCI)
- Author
-
Pegoraro, S, Rizzi, E, Strina, V, Conte, F, Boccolieri, A, Boccolieri, G, Daini, R, Pegoraro, S, Rizzi, E, Strina, V, Conte, F, Boccolieri, A, Boccolieri, G, and Daini, R
- Published
- 2023
6. Teleneuropsychology: normative data for the assessment of memory in online settings
- Author
-
Rizzi, E, Vezzoli, M, Pegoraro, S, Facchin, A, Strina, V, Daini, R, Rizzi, E, Vezzoli, M, Pegoraro, S, Facchin, A, Strina, V, and Daini, R
- Abstract
Introduction: The COVID-19 pandemic has forced significant changes in clinical practice. Psychologists and neuropsychologists had to modify their settings to assess patients’ abilities, switching from an in-person modality to a remote setting by using video calling platforms. Consequently, this change brought about the need for new normative data tailored to remote settings. Aim and methods: The study aimed to develop normative data for the online assessment of neuropsychological memory tests and to compare it with the published norms obtained in standard settings. Two hundred and four healthy Italian volunteers performed three verbal memory tests through the Google Meet platform: the Digit Span (Backward and Forward), the Rey Auditory Verbal Learning, and the Verbal Paired Associated Learning Test. Results: This research provides specific norms that consider the influence of demographic characteristics. Their comparison with published norms shows a medium to high agreement between systems. The present study provides a reference for the clinical use of neuropsychological instruments to assess verbal memory in a remote setting and offers specific recommendations.
- Published
- 2023
7. Effectiveness of cognitive interventions in healthy ageing. A systematic review and meta-analytic approach
- Author
-
Pegoraro, S, Tosi, G, Veronelli, L, Delussi, M, Campo, F, Romano, D, Brattico, E, Daini, R, Romano, DL, Pegoraro, S, Tosi, G, Veronelli, L, Delussi, M, Campo, F, Romano, D, Brattico, E, Daini, R, and Romano, DL
- Published
- 2023
8. Il ruolo delle abilità attentive nel mantenimento della lettura nell’invecchiamento sano
- Author
-
Pegoraro, S, Facchin, A, Luchesa, F, Rolandi, E, Guaita, A, Daini, R, Rolandi, Elena, Pegoraro, S, Facchin, A, Luchesa, F, Rolandi, E, Guaita, A, Daini, R, and Rolandi, Elena
- Published
- 2023
9. Impact of COVID-19 pandemic on healthy olders’ cognitive functioning and the role of cognitive training as a possible protective factor
- Author
-
Pegoraro, S, Rizzi, E, La Rocca, S, Longo, E, Facchin, A, Daini, R, Pegoraro, S, Rizzi, E, La Rocca, S, Longo, E, Facchin, A, and Daini, R
- Subjects
Covid-19, cognitive training, healthy ageing - Published
- 2022
10. Risk Factors for Intracerebral Hemorrhage in Patients with Atrial Fibrillation on Non-Vitamin K Antagonist Oral Anticoagulants for Stroke Prevention
- Author
-
Paciaroni, M. Agnelli, G. Giustozzi, M. Caso, V. Toso, E. Angelini, F. Canavero, I. Micieli, G. Antonenko, K. Rocco, A. Diomedi, M. Katsanos, A.H. Shoamanesh, A. Giannopoulos, S. Ageno, W. Pegoraro, S. Putaala, J. Strbian, D. Sallinen, H. Mac Grory, B.C. Furie, K.L. Stretz, C. Reznik, M.E. Alberti, A. Venti, M. Mosconi, M.G. Vedovati, M.C. Franco, L. Zepponi, G. Romoli, M. Zini, A. Brancaleoni, L. Riva, L. Silvestrelli, G. Ciccone, A. Zedde, M.L. Giorli, E. Kosmidou, M. Ntais, E. Palaiodimou, L. Halvatsiotis, P. Tassinari, T. Saia, V. Ornello, R. Sacco, S. Bandini, F. Mancuso, M. Orlandi, G. Ferrari, E. Pezzini, A. Poli, L. Cappellari, M. Forlivesi, S. Rigatelli, A. Yaghi, S. Scher, E. Frontera, J.A. Masotti, L. Grifoni, E. Caliandro, P. Zauli, A. Reale, G. Marcheselli, S. Gasparro, A. Terruso, V. Arnao, V. Aridon, P. Abdul-Rahim, A.H. Dawson, J. Saggese, C.E. Palmerini, F. Doronin, B. Volodina, V. Toni, D. Risitano, A. Schirinzi, E. Del Sette, M. Lochner, P. Monaco, S. Mannino, M. Tassi, R. Guideri, F. Acampa, M. Martini, G. Lotti, E.M. Padroni, M. Pantoni, L. Rosa, S. Bertora, P. Ntaios, G. Sagris, D. Baldi, A. D'Amore, C. Mumoli, N. Porta, C. Denti, L. Chiti, A. Corea, F. Acciarresi, M. Flomin, Y. Popovic, N. Tsivgoulis, G.
- Subjects
cardiovascular diseases - Abstract
Background and Purpose: Clinical trials on stroke prevention in patients with atrial fibrillation have consistently shown clinical benefit from either warfarin or non-vitamin K antagonist oral anticoagulants (NOACs). NOAC-treated patients have consistently reported to be at lower risk for intracerebral hemorrhage (ICH) than warfarin-treated patients. The aims of this prospective, multicenter, multinational, unmatched, case-control study were (1) to investigate for risk factors that could predict ICH occurring in patients with atrial fibrillation during NOAC treatment and (2) to evaluate the role of CHA2DS2-VASc and HAS-BLED scores in the same setting. Methods: Cases were consecutive patients with atrial fibrillation who had ICH during NOAC treatment. Controls were consecutive patients with atrial fibrillation who did not have ICH during NOAC treatment. As within the CHA2DS2-VASc and HAS-BLED scores there are some risk factors in common, several multivariable logistic regression models were performed to identify independent prespecified predictors for ICH events. Results: Four hundred nineteen cases (mean age, 78.8±8.1 years) and 1526 controls (mean age, 76.0±10.3 years) were included in the study. From the different models performed, independent predictors of ICH were increasing age, concomitant use of antiplatelet agents, active malignancy, high risk of fall, hyperlipidemia, low clearance of creatinine, peripheral artery disease, and white matter changes. Low doses of NOACs (given according to label or not) and congestive heart failure were inversely associated with the risk of ICH. HAS-BLED and CHA2DS2-VASc scores performed poorly in predicting ICH with areas under the curves of 0.496 (95% CI, 0.468-0.525) and 0.530 (95% CI, 0.500-0.560), respectively. Conclusions: Several risk factors were associated to ICH in patients treated with NOACs for stroke prevention but not HAS-BLED and CHA2DS2-VASc scores. © 2021 Lippincott Williams and Wilkins. All rights reserved.
- Published
- 2021
11. Resminostat in advanced hepatocellular carcinoma (HCC) : Overall survival subgroup analysis of prognostic factors in the SHELTER trial
- Author
-
Bitzer, M., Ganten, T. M., Woerns, M. A., Siveke, Jens, Dollinger, M. M., Scheulen, Max Ernst, Wege, H., Giannini, E. G., Cillo, U., Trevisani, F., Santoro, A., Montesarchio, V., Mais, A., Hauns, B., Asche, J., Herz, T., Pegoraro, S., Ammendola, A., Henning, S. W., Hentsch, B., and SHELTER Study Grp
- Subjects
Oncology ,Cancer Research ,medicine.medical_specialty ,business.industry ,Medizin ,Subgroup analysis ,medicine.disease ,chemistry.chemical_compound ,Resminostat ,chemistry ,Hepatocellular carcinoma ,Internal medicine ,medicine ,Overall survival ,business - Abstract
e15088 Background: Previously published results of the phase I/II SHELTER study demonstrated efficacy and safety of the novel pan-HDAC inhibitor resminostat in second-line treatment of HCC patients (pts) who had progressed under first-line sorafenib. As patient baseline characteristics might influence treatment outcome, an analysis of their potential influence on overall survival (OS) was performed. Methods: 45 pts with advanced-stage HCC and centrally confirmed radiologic progression under first-line sorafenib were included in a multi-center, two-arm trial. Resminostat was administered either alone or in combination with sorafenib. A Cox proportional-hazards model was used to evaluate the interaction between baseline characteristics and the effect of the two treatment groups on overall survival. Results: In the combination group, pts with Child-Pugh-A, ECOG 0 or absence of vascular invasion had a statistically significant lower risk of death compared to pts with Child-Pugh-B (HR 0.19, 95% CI 0.06-0.55), ECOG 1 (HR 0.15, 95% CI 0.05-0.44), or vascular invasion (HR 0.37, 95% CI 0.15-0.93), respectively. For pts with BCLC-B there was a strong trend, although not statistically significant, of a lower risk of death when compared to pts with BCLC-C (HR 0.43, 95% CI 0.13-1.49). Etiology, prior TACE therapy, extrahepatic spread and interval between first- and second-line treatment had no impact on overall survival in this study. Similar findings were observed in the monotherapy group. Comparing the impact of these baseline characteristics in the combination and monotherapy group, no statistically significant different influence on OS between both treatment groups was observed. Conclusions: Resminostat in combination with sorafenib provides a substantial OS benefit (median OS of 8.1 months) for advanced HCC patients who had developed progressive tumor disease under first-line sorafenib therapy. Subgroup analysis of patient baseline characteristics revealed a significant influence of Child-Pugh index, ECOG classification, and vascular invasion on overall survival, whereas e.g. the interval between first- and second-line treatment had no impact on overall survival. Clinical trial information: NCT00943449.
- Published
- 2013
12. Peptide Inhibitors of Matrix Metalloproteases
- Author
-
LOUISVILLE UNIV KY, Spatola, A. F., Darlak, K., Pegoraro, S., Nijhawan, K., Anzolin, M., LOUISVILLE UNIV KY, Spatola, A. F., Darlak, K., Pegoraro, S., Nijhawan, K., and Anzolin, M.
- Abstract
Inhibitors of mammalian collagenases and gelatinase have considerable promise for therapy of arthritis, pseudomonas infections, and tumor metastasis. We synthesized effective peptide inhibitors with IC50 values near or below 1 nM against mammalian Type I collagenase and Type IV collagenase (gelatinase). We now focused on (a) demonstration of selectivity vs. other naturally occuring metalloproteases; and (b) improved biostability and oral activity.
- Published
- 1992
13. Sarcolipin, the shorter homologue of phospholamban, forms oligomeric structures in detergent micelles and in liposomes.
- Author
-
Hellstern, S, Pegoraro, S, Karim, C B, Lustig, A, Thomas, D D, Moroder, L, and Engel, J
- Abstract
The human 31-amino acid integral membrane protein sarcolipin (SLN), which regulates the sarcoplasmic reticulum Ca-ATPase in fast-twitch skeletal muscle, was chemically synthesized. Appropriate synthesis and purification strategies were used to achieve high purity and satisfactory yields of this hydrophobic and poorly soluble protein. Structural and functional properties of SLN were analyzed and compared with the homologous region of human phospholamban (PLB) comprising residues Ala(24)-Leu(52) (PLB-(24-52)), the regulatory protein of the cardiac sarcoplasmic reticulum Ca-ATPase. Circular dichroism spectroscopy showed that SLN is a predominantly alpha-helical protein and that the secondary structure is highly resistant to SDS and thermal denaturation. In this respect SLN is remarkably similar to PLB-(24-52). However, SLN is monomeric in SDS gels, whereas PLB-(24-52) shows a monomer-pentamer equilibrium typical for native PLB. Analytical ultracentrifugation experiments revealed that SLN oligomerizes in the presence of the nonionic detergents octylpolyoxyethylene and octyl glucoside in a concentration-dependent manner. No plateau was observed, and a pentameric state was only reached at much higher protein concentrations compared with PLB-(24-52). Chemical cross-linking showed that also in liposomes SLN has the ability to self-associate to oligomers. PLB-(24-52) specifically oligomerized to pentamers in the presence of octylpolyoxyethylene as well as in liposomes at low protein concentrations. In the presence of octylpolyoxyethylene pentamers were the main oligomeric species, whereas in liposomes monomers and dimers were predominant. Increasing the protein concentration led to self-association of PLB-(24-52) pentamers in the presence of octylpolyoxyethylene. Functional reconstitution of Ca-ATPase with PLB-(24-52) and SLN in liposomes showed that both proteins regulate the Ca-ATPase in a similar manner.
- Published
- 2001
- Full Text
- View/download PDF
14. Clinical course of isolated distal deep vein thrombosis in patients with active cancer: a multicenter cohort study
- Author
-
Serena M. Passamonti, Stefano Barco, Giovanni Barillari, Michelangelo Sartori, Federica Bagna, Daniela Mastroiacovo, S. Pegoraro, Walter Ageno, Raffaella Benedetti, Francesco Dentali, Corrado Lodigiani, Nicola Mumoli, Mariasanta Napolitano, M. Di Nisio, M. N. D. Di Minno, Fulvio Pomero, Dentali, F, Pegoraro, S, Barco, S, DI MINNO, Matteo, Mastroiacovo, D, Pomero, F, Lodigiani, C, Bagna, F, Sartori, M, Barillari, G, Mumoli, N, Napolitano, M, Passamonti, S. M, Benedetti, R, Ageno, W, Di Nisio, M., Dentali, F., Pegoraro, S., Barco, S., di Minno, M., Mastroiacovo, D., Pomero, F., Lodigiani, C., Bagna, F., Sartori, M., Barillari, G., Mumoli, N., Napolitano, M., Passamonti, S., Benedetti, R., and Ageno, W.
- Subjects
Male ,medicine.medical_specialty ,distal deep vein thrombosis ,mortality ,neoplasm ,observational study ,venous thromboembolism ,Hematology ,Time Factors ,Deep vein ,Hemorrhage ,030204 cardiovascular system & hematology ,Disease-Free Survival ,03 medical and health sciences ,0302 clinical medicine ,Recurrence ,Risk Factors ,Neoplasms ,Internal medicine ,medicine ,Humans ,030212 general & internal medicine ,Aged ,Proportional Hazards Models ,Retrospective Studies ,Venous Thrombosis ,business.industry ,Incidence ,Clinical course ,Anticoagulants ,Cancer ,distal deep vein thrombosi ,Venous Thromboembolism ,Heparin ,Middle Aged ,medicine.disease ,Thrombosis ,Surgery ,Treatment Outcome ,medicine.anatomical_structure ,Italy ,Female ,Pulmonary Embolism ,business ,Venous thromboembolism ,Distal deep vein thrombosis ,Mortality ,Neoplasm ,Observational study ,medicine.drug ,Cohort study - Abstract
Essentials Isolated distal deep vein thrombosis (IDDVT) is frequently associated with cancer. No study has specifically evaluated the long-term clinical course of cancer-associated IDDVT. Patients with cancer-associated IDDVT are at very high risk of symptomatic recurrence and death. We observed low rates of major bleeding during anticoagulation. SummaryBackground Although isolated distal deep vein thrombosis (IDDVT) is frequently associated with cancer, no study has specifically evaluated the long-term clinical course of IDDVT in this setting. Aim To provide data on the rate of recurrent venous thromboembolism (VTE), major bleeding events and death in IDDVT patients with active cancer. Patients and Methods Consecutive patients with active cancer and an objective IDDVT diagnosis (January 2011 to September 2014) were included from our files. We collected information on baseline characteristics, IDDVT location and extension, VTE risk factors, and type and duration of anticoagulant treatment. Results A total of 308 patients (mean age 66.2 [standard deviation (SD), 13.2 years]; 57.1% female) with symptomatic IDDVT and a solid (n = 261) or hematologic (n = 47) cancer were included at 13 centers. Cancer was metastatic in 148 (48.1%) patients. All but three (99.0%) patients received anticoagulant therapy, which consisted of low-molecular-weight heparin in 288 (93.5%) patients. Vitamin K antagonists were used for the long-term treatment in 46 (14.9%) patients, whereas all others continued the initial parenteral agent for a mean treatment duration of 4.2 months (SD, 4.6 months). During a total follow-up of 355.8 patient-years (mean, 13.9 months), there were 47 recurrent objectively diagnosed VTEs for an incidence rate of 13.2 events per 100 patient-years. During anticoagulant treatment, the annual incidence of major bleeding was 2.0 per 100 patient-years. Conclusions Cancer patients with IDDVT have a high risk of VTE recurrence. Additional studies are warranted to investigate the optimal intensity and duration of anticoagulant treatment for these patients.
- Published
- 2017
- Full Text
- View/download PDF
15. Teleneuropsychology: normative data for the assessment of memory in online settings
- Author
-
Ezia Rizzi, Michela Vezzoli, Sara Pegoraro, Alessio Facchin, Veronica Strina, Roberta Daini, Rizzi, E, Vezzoli, M, Pegoraro, S, Facchin, A, Strina, V, and Daini, R
- Subjects
Psychiatry and Mental health ,M-PSI/03 - PSICOMETRIA ,Verbal Paired Associated Learning ,Digit Span ,Normative data ,Teleneuropsychology ,Neurology (clinical) ,Dermatology ,General Medicine ,M-PSI/02 - PSICOBIOLOGIA E PSICOLOGIA FISIOLOGICA ,Online setting ,Rey Auditory Verbal Learning - Abstract
Introduction The COVID-19 pandemic has forced significant changes in clinical practice. Psychologists and neuropsychologists had to modify their settings to assess patients’ abilities, switching from an in-person modality to a remote setting by using video calling platforms. Consequently, this change brought about the need for new normative data tailored to remote settings. Aim and methods The study aimed to develop normative data for the online assessment of neuropsychological memory tests and to compare it with the published norms obtained in standard settings. Two hundred and four healthy Italian volunteers performed three verbal memory tests through the Google Meet platform: the Digit Span (Backward and Forward), the Rey Auditory Verbal Learning, and the Verbal Paired Associated Learning Test. Results This research provides specific norms that consider the influence of demographic characteristics. Their comparison with published norms shows a medium to high agreement between systems. The present study provides a reference for the clinical use of neuropsychological instruments to assess verbal memory in a remote setting and offers specific recommendations.
- Published
- 2022
16. 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
17. 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
18. 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
19. 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
20. 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
21. 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
22. 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
23. 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
24. Dialogo è comparazione? Saggio sul giudice 'costituzionalista comparatista' nella prospettiva del metodo
- Author
-
CARDUCCI, Michele, L. Pegoraro, S. Bagni, G. Pavani, and Carducci, Michele
- Subjects
Studio delle diverse interpretazioni e giustificazioni fornite al fenomeno del cosiddetto "Judicial Dialogue" e della "Transjudicial Communication" - Abstract
L'articolo analizza le più diffuse teorie sulla emersione di un presunto costituzionalismo globale, inteso come insieme di interpretazioni giurisprudenziali, circolanti attraverso il ricorso alla giurisprudenza straniera.
- Published
- 2014
25. OC-01 - Clinical history of cancer patients with isolated distal deep vein thrombosis: a multicenter cohort study
- Author
-
Corrado Lodigiani, M. N. D. Di Minno, Nicola Mumoli, Daniela Mastroiacovo, Mariasanta Napolitano, M. Di Nisio, S. Pegoraro, Serena M. Passamonti, Walter Ageno, Raffaella Benedetti, Fulvio Pomero, Stefano Barco, Giovanni Barillari, Michelangelo Sartori, Francesco Dentali, F. Bagna, Dentali, F., Pegoraro, S., Barco, S., di Minno, M., Mastroiacovo, D., Pomero, F., Lodigiani, C., Bagna, F., Sartori, M., Barillari, G., Mumoli, N., Napolitano, M., Passamonti, S., Benedetti, R., Ageno, W., and Nisio, M.
- Subjects
medicine.medical_specialty ,medicine.drug_class ,Deep vein ,Population ,Low molecular weight heparin ,030204 cardiovascular system & hematology ,Lower risk ,Gastroenterology ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,medicine ,cancer ,Vein ,education ,Thrombosis history ,distal deep vein thrombosis ,education.field_of_study ,business.industry ,distal deep vein thrombosi ,Hematology ,medicine.disease ,Thrombosis ,Pulmonary embolism ,Surgery ,Venous thrombosis ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,business - Abstract
Introduction Isolated distal deep vein thrombosis (IDDVT) accounts for one-fourth to one-half of all deep vein thrombosis (DVT) of the leg. Patients with IDDVT are frequently treated for a shorter period of time compared to patients with proximal DVT and/or pulmonary embolism (PE) due to a perceived lower risk of recurrence. About 10-20% of patients with venous thromboembolic events (VTEs) have concomitant cancer. Guidelines recommend long-term anticoagulant treatment in this group of patients due to their high risk of VTE recurrence. Unfortunately, information on the clinical history of IDDVT patients is limited and, to date, no study has evaluated the long-term risk of VTE recurrence in IDDVT patients with cancer. Aim To provide information on the clinical history of IDDVT patients with active cancer. Materials and Methods A multicenter, cohort study including active-cancer patients with an objective diagnosis of IDDVT (between January 2011 and September 2014) was conducted. Information on baseline characteristics, thrombosis location and extension, concomitant risk factors, type and duration of treatment was collected. All patients were followed for a minimum of 12 months and up to 24 months. During follow-up, VTE recurrence, major bleeding episodes and death were registered. Potential risk factors for VTE recurrence were evaluated. Results 308 patients (mean age 66.2 ± 13.2 years, female 57.1%) in 13 centers were included, Table 1; 261 patients had solid cancer and 47 patients hematologic cancer. At the time of IDDVT diagnosis, the disease was metastatic in 148 patients (48.1%); 99.0% of patients received anticoagulant treatment: 288 patients (93.5%) were initially treated with low molecular weight heparin, 15 with fondaparinux (5.2%) and 1 with unfractionated heparin; vitamin K antagonists were used in 46 patients (14.9%) only. Total follow-up was 389 patient-years, mean follow-up 15.2 months. Mean duration of treatment was 4.2 months. During the study period there were 47 episodes of VTE recurrence (36 proximal DVT or PE) for a incidence rate of 13.2 events per 100 patient-years; 7 patients had major bleeding (2.3%) and 137 died (44.5%). At multivariate analysis, previous VTE was associated with an increased risk of recurrence (OR 2.10; 95% 1.06, 4.14), whereas patients with gastrointestinal cancer had a lower risk of recurrence (OR 0.26; 95% CI 0.08, 0.86). Table 1 . Baseline characteristics of the population Number of patients 308 Age (years), mean ± SD 66.2 ± 13.2 Women 176 (57.1%) Men 132 (42.9%) Body-mass index > 30 Kg/m 2 25 (8.1%) Concomitant risk factors for VTE Recent surgery or trauma 79 (25.6%) In-patients/immobilization 45 (14.6%) Prolonged bed rest 47 (15.3%) Local or systemic infections 19 (6.2%) Qualifying distal venous thrombosis Axial calf veins 135 (43.8%) Muscolar calf veins 149 (48.4%) Medial gastrocnemius veins 113 (36.7%) Lateral gastrocnemius veins 45 (14.6%) Soleal veins 73 (23.7%) Bilateral venous thrombosis 22 (7.1%) More than one vein involved 127 (41.2%) Previous history of VTE 45 (14.6%) Family history of VTE 16 (5.2%) Primary cancer site Breast 54 (17.5%) Gastrointestinal 51 (16.6%) Pancreas 18 (5.8%) Hepatic 9 (2.9%) Lung 44 (14.3%) Hematologic 47 (15.3%) Prostate 17 (5.5%) Brain 15 (4.9%) Other 53 (17.2%) Metastases 148 (48.1%) Cancer therapy Systemic chemotherapy 174 (56.5%) Radiotherapy 20 (6.5%) Hormonal therapy 36 (11.7%) Anticoagulant therapy 305 (99%) Low-molecular-weight heparin 288 (93.5%) Fondaparinux 16 (5.2%) Unfractionated heparin 1 (0.3%) Heparin-Vitamin K antagonists 46 (14.9%) VTE = venous thromboembolism. Conclusions Cancer patients with IDDVT have a high risk of VTE recurrence. Other studies are warranted to address the adequate management of these patients.
- Published
- 2016
- Full Text
- View/download PDF
26. The proline-rich antimicrobial peptide B7-005: low bacterial resistance, safe for human cells and effective in zebrafish embryo bacteraemia model.
- Author
-
Di Stasi A, Bozzer S, Pacor S, de Pascale L, Morici M, Favero L, Spazzapan M, Pegoraro S, Bulla R, Wilson DN, Macor P, Scocchi M, and Mardirossian M
- Subjects
- Animals, Humans, Drug Resistance, Bacterial drug effects, Disease Models, Animal, Antimicrobial Peptides pharmacology, Antimicrobial Peptides chemistry, Anti-Bacterial Agents pharmacology, Anti-Bacterial Agents therapeutic use, Anti-Bacterial Agents chemistry, Embryo, Nonmammalian drug effects, Proline pharmacology, Proline chemistry, Proline analogs & derivatives, Antimicrobial Cationic Peptides pharmacology, Zebrafish embryology, Bacteremia drug therapy, Bacteremia microbiology, Escherichia coli drug effects, Microbial Sensitivity Tests
- Abstract
Proline-rich antimicrobial peptides (PrAMPs) have gained attention due to their antimicrobial properties and low cytotoxicity. B7-005, a small optimized PrAMP, exhibits a broader spectrum of activity than native PrAMPs, due to an antimicrobial mechanism based on inhibiting prokaryotic protein synthesis and destabilizing bacterial membranes. However, the toxicity and the in vivo efficacy of B7-005 remain poorly understood, so in vitro and in vivo microbiology and toxicology experiments were used to assess its suitability as an anti-infective agent. The incidence of resistance towards B7-005 by E. coli was lower than for other PrAMPs and antibiotics; moreover, it maintained antimicrobial activity in the presence of human serum. B7-005 exerted its antimicrobial effect at a much lower concentration than those causing harmful effects on four different cell types, such as membrane permeabilization or non-lytic depolarization of mitochondria. The latter effect may be related to the inhibition of eukaryotic protein synthesis by B7-005 observed in vitro . In a zebrafish embryo model, B7-005 was well tolerated and reduced mortality from pre-existing E. coli bacteraemia. Overall, B7-005 was safe for human cells and effective against systemic infection in vivo , making it a promising lead for developing new antibiotics.
- Published
- 2024
- Full Text
- View/download PDF
27. Proangiogenic properties of complement protein C1q can contribute to endometriosis.
- Author
-
Agostinis C, Toffoli M, Zito G, Balduit A, Pegoraro S, Spazzapan M, Pascolo L, Romano F, Di Lorenzo G, Mangogna A, Santin A, Spedicati B, Valencic E, Girotto G, Ricci G, Kishore U, and Bulla R
- Subjects
- Humans, Female, Endothelial Cells metabolism, Endothelial Cells immunology, Endometrium immunology, Endometrium metabolism, Endometrium pathology, Macrophages immunology, Macrophages metabolism, Cells, Cultured, Adult, Cell Proliferation, Endometriosis metabolism, Endometriosis immunology, Endometriosis pathology, Endometriosis genetics, Complement C1q genetics, Complement C1q metabolism, Neovascularization, Pathologic genetics, Neovascularization, Pathologic immunology
- Abstract
Endometriosis (EM) is defined as the engraftment and proliferation of functional endometrial-like tissue outside the uterine cavity, leading to a chronic inflammatory condition. While the precise etiology of EM remains elusive, recent studies have highlighted the crucial involvement of a dysregulated immune system. The complement system is one of the predominantly altered immune pathways in EM. Owing to its involvement in the process of angiogenesis, here, we have examined the possible role of the first recognition molecule of the complement classical pathway, C1q. C1q plays seminal roles in several physiological and pathological processes independent of complement activation, including tumor growth, placentation, wound healing, and angiogenesis. Gene expression analysis using the publicly available data revealed that C1q is expressed at higher levels in EM lesions compared to their healthy counterparts. Immunohistochemical analysis confirmed the presence of C1q protein, being localized around the blood vessels in the EM lesions. CD68
+ macrophages are the likely producer of C1q in the EM lesions since cultured EM cells did not produce C1q in vitro . To explore the underlying reasons for increased C1q expression in EM, we focused on its established pro-angiogenic role. Employing various angiogenesis assays on primary endothelial endometriotic cells, such as migration, proliferation, and tube formation assays, we observed a robust proangiogenic effect induced by C1q on endothelial cells in the context of EM. C1q promoted angiogenesis in endothelial cells isolated from EM lesions (as well as healthy ovary that is also rich in C1q). Interestingly, endothelial cells from EM lesions seem to overexpress the receptor for the globular heads of C1q (gC1qR), a putative C1q receptor. Experiments with siRNA to silence gC1qR resulted in diminished capacity of C1q to perform its angiogenic functions, suggesting that C1q is likely to engage gC1qR in the pathophysiology of EM. gC1qR can be a potential therapeutic target in EM patients that will disrupt C1q-mediated proangiogenic activities in EM., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Agostinis, Toffoli, Zito, Balduit, Pegoraro, Spazzapan, Pascolo, Romano, Di Lorenzo, Mangogna, Santin, Spedicati, Valencic, Girotto, Ricci, Kishore and Bulla.)- Published
- 2024
- Full Text
- View/download PDF
28. The Complexity of Reading Revealed by a Study with Healthy Older Adults.
- Author
-
Pegoraro S, Facchin A, Luchesa F, Rolandi E, Guaita A, Arduino LS, and Daini R
- Abstract
Aging, even when healthy, involves changes in cognitive functioning that can gradually affect the everyday activities and well-being of older people. Reading, which requires the integrity of several functions and their integration, is important to maintaining high cognitive and emotional stimulation over time. Our study aimed to investigate whether reading ability declines with aging. To explore also why reading would decline, we explored the changes in the performance of visual and attention tasks. A group of 58 neurologically healthy older people aged from 65 to 75 underwent neuropsychological assessment to investigate their global cognitive functioning, reading skills, crowding, and attention components. We found a decline in reading abilities as a function of aging (β = 0.34, p < 0.05). We did not find an increase in crowding or difficulties in visual acuity. Furthermore, we found no decline with age in tasks of simple reaction times, visuospatial attention, and other single components of attention. Interestingly, we instead found a worsening with age in the Symbol Digit Modalities Test (β = -0.26, p < 0.05), involving attention, working memory, and processing speed, which explains part of the reading decline. Our results suggest that task complexity is a fundamental aspect to account for aging changes.
- Published
- 2024
- Full Text
- View/download PDF
29. Protective role of complement factor H against the development of preeclampsia.
- Author
-
Yasmin H, Agostinis C, Toffoli M, Roy T, Pegoraro S, Balduit A, Zito G, Di Simone N, Ricci G, Madan T, Kishore U, and Bulla R
- Subjects
- Female, Humans, Pregnancy, Complement Factor H metabolism, Endothelial Cells metabolism, Trophoblasts metabolism, Placenta metabolism, Pre-Eclampsia metabolism
- Abstract
Pregnancy is an immunologically regulated, complex process. A tightly controlled complement system plays a crucial role in the successful establishment of pregnancy and parturition. Complement inhibitors at the feto-maternal interface are likely to prevent inappropriate complement activation to protect the fetus. In the present study, we aimed to understand the role of Factor H (FH), a negative regulator of complement activation, in normal pregnancy and in a model of pathological pregnancy, i.e. preeclampsia (PE). The distribution and expression of FH was investigated in placental tissues, various placental cells, and in the sera of healthy (CTRL) or PE pregnant women via immunohistochemistry, RT-qPCR, ELISA, and Western blot. Our results showed a differential expression of FH among the placental cell types, decidual stromal cells (DSCs), decidual endothelial cells (DECs), and extravillous trophoblasts (EVTs). Interestingly, FH was found to be considerably less expressed in the placental tissues of PE patients compared to normal placental tissue both at mRNA and protein levels. Similar results were obtained by measuring circulating FH levels in the sera of third trimester CTRL and PE mothers. Syncytiotrophoblast microvesicles, isolated from the placental tissues of PE and CTRL women, downregulated FH expression by DECs. The present study appears to suggest that FH is ubiquitously present in the normal placenta and plays a homeostatic role during pregnancy., Competing Interests: The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest. The author(s) declared that they were an editorial board member of Frontiers, at the time of submission. This had no impact on the peer review process and the final decision., (Copyright © 2024 Yasmin, Agostinis, Toffoli, Roy, Pegoraro, Balduit, Zito, Di Simone, Ricci, Madan, Kishore and Bulla.)
- Published
- 2024
- Full Text
- View/download PDF
30. Anti-Spike Antibodies Present in the Milk of SARS-CoV-2 Vaccinated Mothers Are Complement-Activating.
- Author
-
Agostinis C, Toffoli M, Balduit A, Mangogna A, Yasmin H, Ragazzon C, Pegoraro S, Campisciano G, Stabile G, Zito G, Kishore U, Comar M, Scrimin F, Bulla R, and Ricci G
- Subjects
- Infant, Newborn, Infant, Female, Humans, COVID-19 Vaccines, Lactation, Milk, Human, Complement System Proteins, Immunoglobulin G, Antibodies, Viral, SARS-CoV-2, COVID-19
- Abstract
Although only 0.8-1% of SARS-CoV-2 infections are in the 0-9 age-group, pneumonia is still the leading cause of infant mortality globally. Antibodies specifically directed against SARS-CoV-2 spike protein (S) are produced during severe COVID-19 manifestations. Following vaccination, specific antibodies are also detected in the milk of breastfeeding mothers. Since antibody binding to viral antigens can trigger activation of the complement classical - pathway, we investigated antibody-dependent complement activation by anti-S immunoglobulins (Igs) present in breast milk following SARS-CoV-2 vaccination. This was in view of the fact that complement could play a fundamentally protective role against SARS-CoV-2 infection in newborns. Thus, 22 vaccinated, lactating healthcare and school workers were enrolled, and a sample of serum and milk was collected from each woman. We first tested for the presence of anti-S IgG and IgA in serum and milk of breastfeeding women by ELISA. We then measured the concentration of the first subcomponents of the three complement pathways (i.e., C1q, MBL, and C3) and the ability of anti-S Igs detected in milk to activate the complement in vitro. The current study demonstrated that vaccinated mothers have anti-S IgG in serum as well as in breast milk, which is capable of activating complement and may confer a protective benefit to breastfed newborns.
- Published
- 2023
- Full Text
- View/download PDF
31. HMGA1 Regulates the Expression of Replication-Dependent Histone Genes and Cell-Cycle in Breast Cancer Cells.
- Author
-
Petrosino S, Pacor S, Pegoraro S, Gazziero VA, Canarutto G, Piazza S, Manfioletti G, and Sgarra R
- Subjects
- Female, Humans, Cell Cycle, Cell Line, Tumor, Epirubicin, Histones metabolism, HMGA1a Protein genetics, HMGA1a Protein metabolism, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- 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 IC
50 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
32. Recurrent Ischemic Stroke and Bleeding in Patients With Atrial Fibrillation Who Suffered an Acute Stroke While on Treatment With Nonvitamin K Antagonist Oral Anticoagulants: The RENO-EXTEND Study.
- Author
-
Paciaroni M, Caso V, Agnelli G, Mosconi MG, Giustozzi M, Seiffge DJ, Engelter ST, Lyrer P, Polymeris AA, Kriemler L, Zietz A, Putaala J, Strbian D, Tomppo L, Michel P, Strambo D, Salerno A, Remillard S, Buehrer M, Bavaud O, Vanacker P, Zuurbier S, Yperzeele L, Loos CMJ, Cappellari M, Emiliani A, Zedde M, Abdul-Rahim A, Dawson J, Cronshaw R, Schirinzi E, Del Sette M, Stretz C, Kala N, Reznik M, Schomer A, Grory BM, Jayaraman M, McTaggart R, Yaghi S, Furie KL, Masotti L, Grifoni E, Toni D, Risitano A, Falcou A, Petraglia L, Lotti EM, Padroni M, Pavolucci L, Lochner P, Silvestrelli G, Ciccone A, Alberti A, Venti M, Traballi L, Urbini C, Kargiotis O, Rocco A, Diomedi M, Marcheselli S, Caliandro P, Zauli A, Reale G, Antonenko K, Rota E, Tassinari T, Saia V, Palmerini F, Aridon P, Arnao V, Monaco S, Cottone S, Baldi A, D'Amore C, Ageno W, Pegoraro S, Ntaios G, Sagris D, Giannopoulos S, Kosmidou M, Ntais E, Romoli M, Pantoni L, Rosa S, Bertora P, Chiti A, Canavero I, Saggese CE, Plocco M, Giorli E, Palaiodimou L, Bakola E, Tsivgoulis G, Bandini F, Gasparro A, Terruso V, Mannino M, Pezzini A, Ornello R, Sacco S, Popovic N, Scoditti U, Genovese A, Denti L, Flomin Y, Mancuso M, Ferrari E, Caselli MC, Ulivi L, Giannini N, and De Marchis GM
- Subjects
- Administration, Oral, Anticoagulants adverse effects, Hemorrhage chemically induced, Hemorrhage complications, Hemorrhage epidemiology, Humans, Prospective Studies, Risk Factors, Atrial Fibrillation chemically induced, Atrial Fibrillation complications, Atrial Fibrillation drug therapy, Brain Ischemia chemically induced, Brain Ischemia drug therapy, Brain Ischemia epidemiology, Ischemic Stroke, Stroke drug therapy, Stroke epidemiology, Stroke prevention & control
- Abstract
Background: In patients with atrial fibrillation who suffered an ischemic stroke while on treatment with nonvitamin K antagonist oral anticoagulants, rates and determinants of recurrent ischemic events and major bleedings remain uncertain., Methods: This prospective multicenter observational study aimed to estimate the rates of ischemic and bleeding events and their determinants in the follow-up of consecutive patients with atrial fibrillation who suffered an acute cerebrovascular ischemic event while on nonvitamin K antagonist oral anticoagulant treatment. Afterwards, we compared the estimated risks of ischemic and bleeding events between the patients in whom anticoagulant therapy was changed to those who continued the original treatment., Results: After a mean follow-up time of 15.0±10.9 months, 192 out of 1240 patients (15.5%) had 207 ischemic or bleeding events corresponding to an annual rate of 13.4%. Among the events, 111 were ischemic strokes, 15 systemic embolisms, 24 intracranial bleedings, and 57 major extracranial bleedings. Predictive factors of recurrent ischemic events (strokes and systemic embolisms) included CHA
2 DS2 -VASc score after the index event (odds ratio [OR], 1.2 [95% CI, 1.0-1.3] for each point increase; P =0.05) and hypertension (OR, 2.3 [95% CI, 1.0-5.1]; P =0.04). Predictive factors of bleeding events (intracranial and major extracranial bleedings) included age (OR, 1.1 [95% CI, 1.0-1.2] for each year increase; P =0.002), history of major bleeding (OR, 6.9 [95% CI, 3.4-14.2]; P =0.0001) and the concomitant administration of an antiplatelet agent (OR, 2.8 [95% CI, 1.4-5.5]; P =0.003). Rates of ischemic and bleeding events were no different in patients who changed or not changed the original nonvitamin K antagonist oral anticoagulants treatment (OR, 1.2 [95% CI, 0.8-1.7])., Conclusions: Patients suffering a stroke despite being on nonvitamin K antagonist oral anticoagulant therapy are at high risk of recurrent ischemic stroke and bleeding. In these patients, further research is needed to improve secondary prevention by investigating the mechanisms of recurrent ischemic stroke and bleeding.- Published
- 2022
- Full Text
- View/download PDF
33. HMGA1 positively regulates the microtubule-destabilizing protein stathmin promoting motility in TNBC cells and decreasing tumour sensitivity to paclitaxel.
- Author
-
Sgubin M, Pegoraro S, Pellarin I, Ros G, Sgarra R, Piazza S, Baldassarre G, Belletti B, and Manfioletti G
- Subjects
- HMGA1a Protein genetics, HMGA1a Protein metabolism, Humans, Microtubules metabolism, Neoplasm Recurrence, Local metabolism, Paclitaxel pharmacology, Paclitaxel therapeutic use, Stathmin genetics, Stathmin metabolism, Triple Negative Breast Neoplasms drug therapy, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms metabolism
- 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 p27
kip1 (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., (© 2022. The Author(s).)- Published
- 2022
- Full Text
- View/download PDF
34. Temporally distinct post-replicative repair mechanisms fill PRIMPOL-dependent ssDNA gaps in human cells.
- Author
-
Tirman S, Quinet A, Wood M, Meroni A, Cybulla E, Jackson J, Pegoraro S, Simoneau A, Zou L, and Vindigni A
- Subjects
- Antineoplastic Agents pharmacology, BRCA1 Protein genetics, BRCA1 Protein metabolism, BRCA2 Protein metabolism, Cell Line, Tumor, DNA Helicases genetics, DNA Helicases metabolism, DNA Primase genetics, DNA, Neoplasm genetics, DNA-Binding Proteins genetics, DNA-Binding Proteins metabolism, DNA-Directed DNA Polymerase genetics, Genomic Instability, HEK293 Cells, Humans, MRE11 Homologue Protein genetics, MRE11 Homologue Protein metabolism, Multifunctional Enzymes genetics, Neoplasms drug therapy, Neoplasms genetics, Neoplasms pathology, Nucleotidyltransferases genetics, Nucleotidyltransferases metabolism, Proliferating Cell Nuclear Antigen genetics, Proliferating Cell Nuclear Antigen metabolism, Time Factors, Ubiquitin-Conjugating Enzymes genetics, Ubiquitin-Conjugating Enzymes metabolism, Ubiquitin-Protein Ligases genetics, Ubiquitin-Protein Ligases metabolism, Ubiquitination, DNA Breaks, Single-Stranded, DNA Primase metabolism, DNA Repair, DNA Replication, DNA, Neoplasm biosynthesis, DNA-Directed DNA Polymerase metabolism, G2 Phase, Multifunctional Enzymes metabolism, Neoplasms metabolism, S Phase
- Abstract
PRIMPOL repriming allows DNA replication to skip DNA lesions, leading to ssDNA gaps. These gaps must be filled to preserve genome stability. Using a DNA fiber approach to directly monitor gap filling, we studied the post-replicative mechanisms that fill the ssDNA gaps generated in cisplatin-treated cells upon increased PRIMPOL expression or when replication fork reversal is defective because of SMARCAL1 inactivation or PARP inhibition. We found that a mechanism dependent on the E3 ubiquitin ligase RAD18, PCNA monoubiquitination, and the REV1 and POLζ translesion synthesis polymerases promotes gap filling in G2. The E2-conjugating enzyme UBC13, the RAD51 recombinase, and REV1-POLζ are instead responsible for gap filling in S, suggesting that temporally distinct pathways of gap filling operate throughout the cell cycle. Furthermore, we found that BRCA1 and BRCA2 promote gap filling by limiting MRE11 activity and that simultaneously targeting fork reversal and gap filling enhances chemosensitivity in BRCA-deficient cells., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
35. Gene network analysis using SWIM reveals interplay between the transcription factor-encoding genes HMGA1, FOXM1, and MYBL2 in triple-negative breast cancer.
- Author
-
Fiscon G, Pegoraro S, Conte F, Manfioletti G, and Paci P
- Subjects
- Atlases as Topic, Carcinoma, Ductal, Breast metabolism, Carcinoma, Ductal, Breast pathology, Cell Cycle Proteins metabolism, Cell Line, Tumor, Data Mining, Databases, Genetic, Datasets as Topic, Female, Forkhead Box Protein M1 metabolism, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, HMGA1a Protein metabolism, Humans, Multigene Family, Protein Binding, Signal Transduction, Trans-Activators metabolism, Triple Negative Breast Neoplasms metabolism, Triple Negative Breast Neoplasms pathology, Carcinoma, Ductal, Breast genetics, Cell Cycle Proteins genetics, Forkhead Box Protein M1 genetics, Gene Regulatory Networks, HMGA1a Protein genetics, Trans-Activators genetics, Triple Negative Breast Neoplasms genetics
- 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., (© 2021 Federation of European Biochemical Societies.)
- Published
- 2021
- Full Text
- View/download PDF
36. Risk Factors for Intracerebral Hemorrhage in Patients With Atrial Fibrillation on Non-Vitamin K Antagonist Oral Anticoagulants for Stroke Prevention.
- Author
-
Paciaroni M, Agnelli G, Giustozzi M, Caso V, Toso E, Angelini F, Canavero I, Micieli G, Antonenko K, Rocco A, Diomedi M, Katsanos AH, Shoamanesh A, Giannopoulos S, Ageno W, Pegoraro S, Putaala J, Strbian D, Sallinen H, Mac Grory BC, Furie KL, Stretz C, Reznik ME, Alberti A, Venti M, Mosconi MG, Vedovati MC, Franco L, Zepponi G, Romoli M, Zini A, Brancaleoni L, Riva L, Silvestrelli G, Ciccone A, Zedde ML, Giorli E, Kosmidou M, Ntais E, Palaiodimou L, Halvatsiotis P, Tassinari T, Saia V, Ornello R, Sacco S, Bandini F, Mancuso M, Orlandi G, Ferrari E, Pezzini A, Poli L, Cappellari M, Forlivesi S, Rigatelli A, Yaghi S, Scher E, Frontera JA, Masotti L, Grifoni E, Caliandro P, Zauli A, Reale G, Marcheselli S, Gasparro A, Terruso V, Arnao V, Aridon P, Abdul-Rahim AH, Dawson J, Saggese CE, Palmerini F, Doronin B, Volodina V, Toni D, Risitano A, Schirinzi E, Del Sette M, Lochner P, Monaco S, Mannino M, Tassi R, Guideri F, Acampa M, Martini G, Lotti EM, Padroni M, Pantoni L, Rosa S, Bertora P, Ntaios G, Sagris D, Baldi A, D'Amore C, Mumoli N, Porta C, Denti L, Chiti A, Corea F, Acciarresi M, Flomin Y, Popovic N, and Tsivgoulis G
- Subjects
- Administration, Oral, Aged, Aged, 80 and over, Case-Control Studies, Female, Humans, Male, Middle Aged, Prospective Studies, Risk Factors, Stroke etiology, Antithrombins therapeutic use, Atrial Fibrillation complications, Cerebral Hemorrhage chemically induced, Stroke prevention & control
- Abstract
[Figure: see text].
- Published
- 2021
- Full Text
- View/download PDF
37. On the importance of primary and community healthcare in relation to global health and environmental threats: lessons from the COVID-19 crisis.
- Author
-
Lauriola P, Martín-Olmedo P, Leonardi GS, Bouland C, Verheij R, Dückers MLA, van Tongeren M, Laghi F, van den Hazel P, Gokdemir O, Segredo E, Etzel RA, Abelsohn A, Bianchi F, Romizi R, Miserotti G, Romizi F, Bortolotti P, Vinci E, Giustetto G, Santamaria M, Serafini A, Pegoraro S, Agius R, and Zeka A
- Subjects
- Climate Change, Education, Global Health, Humans, Politics, SARS-CoV-2, Socioeconomic Factors, COVID-19 epidemiology, Community Health Services, Public Health
- Abstract
In the course of the COVID-19 pandemic, it has become clear that primary healthcare systems play a critical role in clinical care, such as patient screening, triage, physical and psychological support and also in promoting good community advice and awareness in coordination with secondary healthcare and preventive care. Because of the role of social and environmental factors in COVID-19 transmission and burden of disease, it is essential to ensure that there is adequate coordination of population-based health services and public health interventions. The COVID-19 pandemic has shown the primary and community healthcare (P&CHC) system's weaknesses worldwide. In many instances, P&CHC played only a minor role, the emphasis being on hospital and intensive care beds. This was compounded by political failures, in supporting local community resilience. Placing community building, social cohesion and resilience at the forefront of dealing with the COVID-19 crisis can help align solutions that provide a vision of 'planetary health'. This can be achieved by involving local well-being and participation in the face of any pervasive health and environmental crisis, including other epidemics and large-scale ecological crises. This paper proposes that P&CHC should take on two critical roles: first, to support local problem-solving efforts and to serve as a partner in innovative approaches to safeguarding community well-being; and second, to understand the local environment and health risks in the context of the global health perspective. We see this as an opportunity of immediate value and broad consequence beyond the control of the COVID-19 pandemic., Competing Interests: Competing interests: None declared., (© Author(s) (or their employer(s)) 2021. Re-use permitted under CC BY-NC. No commercial re-use. See rights and permissions. Published by BMJ.)
- Published
- 2021
- Full Text
- View/download PDF
38. High Mobility Group A (HMGA): Chromatin Nodes Controlled by a Knotty miRNA Network.
- Author
-
Sgarra R, Pegoraro S, D'Angelo D, Ros G, Zanin R, Sgubin M, Petrosino S, Battista S, and Manfioletti G
- Subjects
- Animals, Gene Expression Regulation, Neoplastic genetics, Humans, Neoplasms genetics, Chromatin genetics, HMGA Proteins genetics, MicroRNAs genetics
- 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., Competing Interests: The authors declare no conflicts of interest.
- Published
- 2020
- Full Text
- View/download PDF
39. HMGA2 Antisense Long Non-coding RNAs as New Players in the Regulation of HMGA2 Expression and Pancreatic Cancer Promotion.
- Author
-
Ros G, Pegoraro S, De Angelis P, Sgarra R, Zucchelli S, Gustincich S, and Manfioletti G
- 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., (Copyright © 2020 Ros, Pegoraro, De Angelis, Sgarra, Zucchelli, Gustincich and Manfioletti.)
- Published
- 2020
- Full Text
- View/download PDF
40. HMGA1 Modulates Gene Transcription Sustaining a Tumor Signalling Pathway Acting on the Epigenetic Status of Triple-Negative Breast Cancer Cells.
- Author
-
Penzo C, Arnoldo L, Pegoraro S, Petrosino S, Ros G, Zanin R, Wiśniewski JR, Manfioletti G, and Sgarra R
- 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., Competing Interests: The authors declare no conflict of interest.
- Published
- 2019
- Full Text
- View/download PDF
41. HMGA1 promotes breast cancer angiogenesis supporting the stability, nuclear localization and transcriptional activity of FOXM1.
- Author
-
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
- Animals, Cell Line, Tumor, Culture Media, Conditioned pharmacology, Endothelial Cells cytology, Endothelial Cells drug effects, Endothelial Cells metabolism, Female, Forkhead Box Protein M1 chemistry, Forkhead Box Protein M1 genetics, Gene Expression Profiling, Gene Expression Regulation, Neoplastic, Gene Silencing, HEK293 Cells, HMGA1a Protein genetics, Humans, Prognosis, Promoter Regions, Genetic, Protein Stability, Sequence Analysis, RNA, Survival Analysis, Transcription, Genetic, Triple Negative Breast Neoplasms metabolism, Zebrafish, Cell Nucleus metabolism, Forkhead Box Protein M1 metabolism, HMGA1a Protein metabolism, Triple Negative Breast Neoplasms genetics, Vascular Endothelial Growth Factor A genetics
- 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
42. The High Mobility Group A1 (HMGA1) Chromatin Architectural Factor Modulates Nuclear Stiffness in Breast Cancer Cells.
- Author
-
Senigagliesi B, Penzo C, Severino LU, Maraspini R, Petrosino S, Morales-Navarrete H, Pobega E, Ambrosetti E, Parisse P, Pegoraro S, Manfioletti G, Casalis L, and Sgarra R
- Subjects
- Breast Neoplasms genetics, Breast Neoplasms mortality, Breast Neoplasms pathology, Cell Cycle genetics, Cell Line, Tumor, Chromatin genetics, Chromatin metabolism, Female, Gene Expression, HMGA Proteins genetics, Histones metabolism, Humans, Kaplan-Meier Estimate, Phosphorylation, Prognosis, Protein Binding, Breast Neoplasms metabolism, Cell Nucleus metabolism, HMGA Proteins metabolism
- 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
43. Proneural-Mesenchymal Transition: Phenotypic Plasticity to Acquire Multitherapy Resistance in Glioblastoma.
- Author
-
Fedele M, Cerchia L, Pegoraro S, Sgarra R, and Manfioletti G
- Subjects
- Animals, Biomarkers, Combined Modality Therapy, Drug Resistance, Neoplasm, Epithelial-Mesenchymal Transition, Glioblastoma therapy, Humans, Neoplastic Stem Cells metabolism, Signal Transduction, Treatment Outcome, Tumor Microenvironment, Cell Transformation, Neoplastic, Glioblastoma etiology, Glioblastoma pathology, Phenotype
- Abstract
Glioblastoma (GBM) is an extremely aggressive tumor of the central nervous system, with a prognosis of 12-15 months and just 3-5% of survival over 5 years. This is mainly because most patients suffer recurrence after treatment that currently consists in maximal resection followed by radio- and chemotherapy with temozolomide. The recurrent tumor shows a more aggressive behavior due to a phenotypic shift toward the mesenchymal subtype. Proneural-mesenchymal transition (PMT) may represent for GBM the equivalent of epithelial-mesenchymal transition associated with other aggressive cancers. In this review we frame this process in the high degree of phenotypic inter- and intra-tumor heterogeneity of GBM, which exists in different subtypes, each one characterized by further phenotypic variability in its stem-cell compartment. Under the selective pressure of different treatment agents PMT is induced. The mechanisms involved, as well as the significance of such event in the acquisition of a multitherapy resistance phenotype, are taken in consideration for future perspectives in new anti-GBM therapeutic options.
- Published
- 2019
- Full Text
- View/download PDF
44. Transcriptional Regulation of Glucose Metabolism: The Emerging Role of the HMGA1 Chromatin Factor.
- Author
-
Chiefari E, Foti DP, Sgarra R, Pegoraro S, Arcidiacono B, Brunetti FS, Greco M, Manfioletti G, and Brunetti A
- 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
45. Long-term recurrence of venous thromboembolism after short-term treatment of symptomatic isolated distal deep vein thrombosis: A cohort study.
- Author
-
Donadini MP, Dentali F, Pegoraro S, Pomero F, Brignone C, Guasti L, Steidl L, and Ageno W
- Subjects
- Adult, Aged, Aged, 80 and over, Disease-Free Survival, Drug Administration Schedule, Female, Humans, Kaplan-Meier Estimate, Male, Middle Aged, Recurrence, Retrospective Studies, Risk Factors, Time Factors, Treatment Outcome, Venous Thromboembolism diagnostic imaging, Venous Thrombosis diagnostic imaging, Young Adult, Anticoagulants administration & dosage, Heparin, Low-Molecular-Weight administration & dosage, Venous Thromboembolism drug therapy, Venous Thrombosis drug therapy
- Abstract
Isolated distal deep vein thrombosis (IDDVT) is a common clinical manifestation of venous thromboembolism (VTE). However, there are only scant and heterogeneous data available on the long-term risk of recurrent VTE after IDDVT, and the optimal therapeutic management remains uncertain. We carried out a retrospective cohort study of consecutive patients diagnosed with symptomatic IDDVT between 2004 and 2011, according to a predefined short-term treatment protocol (low molecular weight heparin (LMWH) for 4-6 weeks). The primary outcome was the occurrence of recurrent VTE. A total of 321 patients were enrolled. IDDVT was associated with a transient risk factor or cancer in 165 (51.4%) and 56 (17.4%) patients, respectively. LMWH was administered for 4-6 weeks to 280 patients (87.2%), who were included in the primary analysis. Overall, during a mean follow-up of 42.3 months, 42 patients (15%) developed recurrent VTE, which occurred as proximal DVT or PE in 21 cases. The recurrence rate of VTE per 100 patient-years was 3.5 in patients with transient risk factors, 7.2 in patients with unprovoked IDDVT, and 5.9 in patients with cancer ( p=0.018). At multivariable analysis, unprovoked IDDVT and previous VTE were significantly associated with recurrent VTE (HR 2.16, 95% CI 1.12-4.16 and HR 1.97, 95% CI 1.01-3.86, respectively). In conclusion, the long-term risk of recurrent VTE after IDDVT treated for 4-6 weeks is not negligible, in particular in patients with unprovoked IDDVT or cancer. Further studies are needed to clarify whether a longer, but definite treatment duration effectively prevents these recurrences.
- Published
- 2017
- Full Text
- View/download PDF
46. HMGA1 regulates the Plasminogen activation system in the secretome of breast cancer cells.
- Author
-
Resmini G, Rizzo S, Franchin C, Zanin R, Penzo C, Pegoraro S, Ciani Y, Piazza S, Arrigoni G, Sgarra R, and Manfioletti G
- Subjects
- Cell Line, Tumor, Female, HEK293 Cells, Humans, Neoplasm Proteins genetics, Plasminogen genetics, Triple Negative Breast Neoplasms genetics, Triple Negative Breast Neoplasms pathology, Cell Transformation, Neoplastic, Gene Expression Regulation, Neoplastic, HMGA Proteins metabolism, Neoplasm Proteins metabolism, Plasminogen metabolism, Triple Negative Breast Neoplasms metabolism
- 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
- Full Text
- View/download PDF
47. Erratum: SC83288 is a clinical development candidate for the treatment of severe malaria.
- Author
-
Pegoraro S, Duffey M, Otto TD, Wang Y, Rösemann R, Baumgartner R, Fehler SK, Lucantoni L, Avery VM, Moreno-Sabater A, Mazier D, Vial HJ, Strobl S, Sanchez CP, and Lanzer M
- Published
- 2017
- Full Text
- View/download PDF
48. SC83288 is a clinical development candidate for the treatment of severe malaria.
- Author
-
Pegoraro S, Duffey M, Otto TD, Wang Y, Rösemann R, Baumgartner R, Fehler SK, Lucantoni L, Avery VM, Moreno-Sabater A, Mazier D, Vial HJ, Strobl S, Sanchez CP, and Lanzer M
- Subjects
- Acute Disease, Animals, Antimalarials chemical synthesis, Antimalarials pharmacokinetics, Calcium-Transporting ATPases genetics, Calcium-Transporting ATPases metabolism, Disease Models, Animal, Drug Resistance, Endoplasmic Reticulum metabolism, Gene Expression, Humans, Inhibitory Concentration 50, Ion Transport, Malaria, Falciparum parasitology, Male, Mice, Mice, Inbred NOD, Mice, SCID, Plasmodium falciparum genetics, Plasmodium falciparum growth & development, Plasmodium falciparum metabolism, Structure-Activity Relationship, Antimalarials pharmacology, Calcium-Transporting ATPases antagonists & inhibitors, Endoplasmic Reticulum drug effects, Malaria, Falciparum drug therapy, Plasmodium falciparum drug effects
- Abstract
Severe malaria is a life-threatening complication of an infection with the protozoan parasite Plasmodium falciparum, which requires immediate treatment. Safety and efficacy concerns with currently used drugs accentuate the need for new chemotherapeutic options against severe malaria. Here we describe a medicinal chemistry program starting from amicarbalide that led to two compounds with optimized pharmacological and antiparasitic properties. SC81458 and the clinical development candidate, SC83288, are fast-acting compounds that can cure a P. falciparum infection in a humanized NOD/SCID mouse model system. Detailed preclinical pharmacokinetic and toxicological studies reveal no observable drawbacks. Ultra-deep sequencing of resistant parasites identifies the sarco/endoplasmic reticulum Ca
2+ transporting PfATP6 as a putative determinant of resistance to SC81458 and SC83288. Features, such as fast parasite killing, good safety margin, a potentially novel mode of action and a distinct chemotype support the clinical development of SC83288, as an intravenous application for the treatment of severe malaria., Competing Interests: The authors declare no competing financial interests.- Published
- 2017
- Full Text
- View/download PDF
49. The Architectural Chromatin Factor High Mobility Group A1 Enhances DNA Ligase IV Activity Influencing DNA Repair.
- Author
-
Pellarin I, Arnoldo L, Costantini S, Pegoraro S, Ros G, Penzo C, Triolo G, Demarchi F, Sgarra R, Vindigni A, and Manfioletti G
- Subjects
- Cell Line, Tumor, Chromatin metabolism, Chromatography, High Pressure Liquid, Comet Assay, HMGA1a Protein genetics, HMGA2 Protein genetics, HMGA2 Protein metabolism, Histones metabolism, Humans, Ku Autoantigen metabolism, MCF-7 Cells, Microscopy, Fluorescence, Phosphorylation, Recombinant Proteins biosynthesis, Recombinant Proteins chemistry, Recombinant Proteins isolation & purification, Substrate Specificity, Chromatin chemistry, DNA Ligase ATP metabolism, DNA Repair, HMGA1a Protein metabolism
- 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., Competing Interests: The authors have declared that no competing interests exist.
- Published
- 2016
- Full Text
- View/download PDF
50. Hmga2 is required for neural crest cell specification in Xenopus laevis.
- Author
-
Macrì S, Simula L, Pellarin I, Pegoraro S, Onorati M, Sgarra R, Manfioletti G, and Vignali R
- Subjects
- Animals, Cell Line, Tumor, Cell Movement genetics, Female, Gene Expression Regulation, Developmental, Gene Regulatory Networks genetics, HMGA2 Protein genetics, MSX1 Transcription Factor genetics, Mammary Neoplasms, Experimental genetics, Mammary Neoplasms, Experimental metabolism, Mammary Neoplasms, Experimental pathology, Mice, Morpholinos genetics, Neural Crest cytology, PAX3 Transcription Factor, Paired Box Transcription Factors genetics, RNA Interference, RNA, Small Interfering genetics, Transcription Factors genetics, Transforming Growth Factor beta metabolism, Xenopus Proteins genetics, Cell Differentiation genetics, Epithelial-Mesenchymal Transition genetics, HMGA2 Protein physiology, Neural Crest embryology, Xenopus Proteins physiology, Xenopus laevis embryology
- Abstract
HMGA proteins are small nuclear proteins that bind DNA by conserved AT-hook motifs, modify chromatin architecture and assist in gene expression. Two HMGAs (HMGA1 and HMGA2), encoded by distinct genes, exist in mammals and are highly expressed during embryogenesis or reactivated in tumour progression. We here addressed the in vivo role of Xenopus hmga2 in the neural crest cells (NCCs). We show that hmga2 is required for normal NCC specification and development. hmga2 knockdown leads to severe disruption of major skeletal derivatives of anterior NCCs. We show that, within the NCC genetic network, hmga2 acts downstream of msx1, and is required for msx1, pax3 and snail2 activities, thus participating at different levels of the network. Because of hmga2 early effects in NCC specification, the subsequent epithelial-mesenchymal transition (EMT) and migration of NCCs towards the branchial pouches are also compromised. Strictly paralleling results on embryos, interfering with Hmga2 in a breast cancer cell model for EMT leads to molecular effects largely consistent with those observed on NCCs. These data indicate that Hmga2 is recruited in key molecular events that are shared by both NCCs and tumour cells., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.