46 results on '"Foglio E"'
Search Results
2. Fatigue Characterization and Optimization of the Production Process of Heavy Section Ductile Iron Castings
- Author
-
Foglio, E., Gelfi, M., Pola, A., Goffelli, S., and Lusuardi, D.
- Published
- 2017
- Full Text
- View/download PDF
3. Mechanical characterization of heavy section ductile cast iron components
- Author
-
Foglio, E., Gelfi, M., Pola, A., and Lusuardi, D.
- Subjects
Chunky graphite ,Ductile cast iron ,Fatigue ,Fracture analysis ,Microporosity ,Spiky graphite ,Condensed Matter Physics ,Mechanics of Materials ,2506 - Published
- 2019
4. SIRT1-SIRT3 Axis Regulates Cellular Response to Oxidative Stress and Etoposide
- Author
-
Carnevale, I., Pellegrini, L., D'Aquila, P., Saladini, S., Lococo, E., Polletta, L., Vernucci, E., Foglio, E., and Coppola, S.
- Published
- 2017
5. Effect of Shrinkage Porosity and Degenerated Graphite on Fatigue Crack Initiation in Ductile Cast Iron
- Author
-
Foglio, E., primary, Gelfi, Marcello, additional, Pola, Annalisa, additional, and Lusuardi, D., additional
- Published
- 2017
- Full Text
- View/download PDF
6. Fatigue Characterization and Optimization of the Production Process of Heavy Section Ductile Iron Castings
- Author
-
Foglio, E., primary, Gelfi, M., additional, Pola, A., additional, Goffelli, S., additional, and Lusuardi, D., additional
- Published
- 2016
- Full Text
- View/download PDF
7. Silicic acid in drinking water modulates nitric oxide and aquaporin in vessels
- Author
-
Rodella, Luigi Fabrizio, Favero, Gaia, Foglio, E, Merigo, D, Labanca, M, and Rezzani, Rita
- Published
- 2012
8. Muscle LIM protein/CSRP3: a mechanosensor with a role in autophagy
- Author
-
Rashid, M M, primary, Runci, A, additional, Polletta, L, additional, Carnevale, I, additional, Morgante, E, additional, Foglio, E, additional, Arcangeli, T, additional, Sansone, L, additional, Russo, M A, additional, and Tafani, M, additional
- Published
- 2015
- Full Text
- View/download PDF
9. ANALYSIS OF THE BENEFICIAL EFFECTS OF TAURINE AGAINST PUROMYCIN-INDUCED NEPHROSIS IN RATS
- Author
-
Stacchiotti, Alessandra, Foglio, E., Lavazza, A., Rodella, Luigi Fabrizio, and Rezzani, Rita
- Subjects
kidney ,TEM ,puromycin - Published
- 2010
10. Silicon in drinking water prevents loss of nitroxidergic neurons in brain
- Author
-
Buffoli, Barbara, Foglio, E, Labanca, M, Bonomini, Francesca, and Rodella, Luigi Fabrizio
- Published
- 2009
11. Aluminium exposure induces Alzheimer's disease-like histopathological alterations in mouse brain
- Author
-
Rodella, L. F., Ricci, F., Elisa Borsani, Stacchiotti, A., Foglio, E., Favero, G., Rezzani, R., Mariani, C., and Bianchi, R.
- Subjects
Male ,Plaque, Amyloid ,Cerebellar Cortex ,Mice ,Alzheimer Disease ,alzheimer disease ,mouse ,brain ,aluminium ,Animals ,Humans ,Toxicity Tests, Chronic ,Endoplasmic Reticulum Chaperone BiP ,Heat-Shock Proteins ,Neurons ,616.8- Neurología. Neuropatología. Sistema Nervioso ,Amyloid beta-Peptides ,Aluminium sulphate ,Brain ,Immunohistochemistry ,Mice, Inbred C57BL ,Disease Models, Animal ,Alzheimer's disease ,Beta amyloid ,GRP78 ,Neurodegenerative damage ,Settore MED/26 - Neurologia ,Alzheimer’s disease ,Aluminum ,Molecular Chaperones - Abstract
Aluminium (Al) is a neurotoxic metal and Al exposure may be a factor in the aetiology of various neurodegenerative diseases such as Alzheimer’s disease (AD). The major pathohistological findings in the AD brain are the presence of neuritic plaques containing ßamyloid (Aß) which may interfere with neuronal communication. Moreover, it has been observed that GRP78, a stress-response protein induced by conditions that adversely affect endoplasmic reticulum (ER) function, is reduced in the brain of AD patients. In this study, we investigated the correlation between the expression of Aß and GRP78 in the brain cortex of mice chronically treated with aluminium sulphate. Chronic exposure over 12 months to aluminium sulphate in drinking water resulted in deposition of Aß similar to that seen in congophilic amyloid angiopathy (CAA) in humans and a reduction in neuronal expression of GRP78 similar to what has previously been observed in Alzheimer’s disease. So, we hypothesise that chronic Al administration is responsible for oxidative cell damage that interferes with ER functions inducing Aß accumulation and neurodegenerative damage.
- Published
- 2008
12. Hepatotoxic effects in mouse by prolonged exposure to aluminium
- Author
-
Stacchiotti, Alessandra, Foglio, E, Pedretti, N, Lavazza, A, Rezzani, Rita, Bianchi, R, and Rodella, Luigi Fabrizio
- Published
- 2007
13. Differential stress proteins expression in NRK-52E cells exposed to Hg(II) or Pb(II)
- Author
-
Stacchiotti, Alessandra, Morandini, F., Bettoni, F., Foglio, E., Cadei, M., Rodella, Luigi Fabrizio, Grigolato, Pier Giovanni, and Aleo, Maria Francesca
- Published
- 2007
14. Taurine limits acute puromycin-induced nephrosis
- Author
-
Stacchiotti, Alessandra, Ricci, E, Foglio, E, Rezzani, Rita, Rodella, Luigi Fabrizio, and Bianchi, R.
- Published
- 2006
15. Regular consumption of a silicic acid-rich water prevents aluminium-induced alterations of nitrergic neurons in mouse brain: histochemical and immunohistochemical studies
- Author
-
Foglio, E., Buffoli, B., Christopher Exley, Rezzani, R., and Rodella, L. F.
- Subjects
inorganic chemicals ,Cerebral Cortex ,Male ,Silicon ,Drinking Water ,Silicic Acid ,Drinking ,NADPH Dehydrogenase ,Brain ,Nitric Oxide Synthase Type I ,respiratory system ,Aluminium ,Nitrergic neurons ,complex mixtures ,Mice, Inbred C57BL ,Mice ,Neuroprotective Agents ,Nitrergic Neurons ,6 - Ciencias aplicadas::61 - Medicina::611 - Anatomía [CDU] ,Alum Compounds ,Animals ,Mineral Waters ,Toxicity Tests, Chronic ,Drug Antagonism ,Water Pollutants, Chemical - Abstract
Silicon is not generally considered an essential nutrient for mammals and, to date, whether it has a biological role or beneficial effects in humans is not known. The results of a number of studies suggest that dietary silicon supplementation might have a protective effect both for limiting aluminium absorption across the gut and for the removal of systemic aluminium via the urine, hence, preventing potential accumulation of aluminium in the brain. Since our previous studies demonstrated that aluminium exposure reduces the number of nitrergic neurons, the aim of the present study was to compare the distribution and the morphology of NO-containing neurons in brain cortex of mice exposed to aluminium sulphate dissolved in silicic acid-rich or poor drinking water to assess the potential protective role of silicon against aluminium toxicity in the brain. NADPH-d histochemistry and nNOS immunohistochemistry showed that high concentrations of silicon in drinking water were able to minimize the impairment of the function of nitrergic neurons induced by aluminium administration. We found that silicon protected against aluminium-induced damage to the nitrergic system: in particular, we demonstrated that silicon maintains the number of nitrergic neurons and their expression of nitrergic enzymes at physiological levels, even after a 12 and 15 month exposure to aluminium.
16. Cardiomyocyte Regeneration in Human Myocarditis.
- Author
-
Frustaci A, Foglio E, Limana F, Magnocavallo M, Frustaci E, Lupacchini L, and Verardo R
- Abstract
Background: Newly generated cardiomyocytes (NGCs) concur with the recovery of human myocarditis occurring spontaneously in around 50% of cases. However, NGCs decline with age, and their modality of myocardial homing and integration are still unclear., Methods: We retrospectively assessed NGCs in 213 consecutive patients with endomyocardial biopsy denoting acute myocarditis, with normal coronaries and valves. Tissue samples were processed for histology (H&E), immunohistochemistry for the evaluation of inflammatory infiltrates, immunostaining for alpha-sarcomeric-actin, junctional connexin-43, Ki-67, and phosphorylated STAT3 (p-STAT3), and Western blot (WB) for HMGB1. Frozen samples were analyzed using polymerase chain reaction (PCR) for cardiotropic viruses. Controls included 20 normal surgical biopsies., Results: NGCs were defined as small myocytes (diameter < 10 µm) with nuclear positivity to Ki-67 and p-STAT3 and positive immunostaining for cytoplasmic α-sarcomeric actin and connexin-43. Their number/mm
2 in relation to age and pathway of integration was evaluated. NGCs crossed the membrane and grew integrated within the empty necrotic myocytes. NGC mean diameter was 6.6 ± 3.34 vs. 22.5 ± 3.11 µm adult cells; their number, in comparison to LVEF, was 86.3 ± 10.3/mm2 in patients between 18 and 40 years, 50.4 ± 13.8/mm2 in those between 41 and 60, and 15.1 ± 5.7/mm2 in those between 61 and 80. Control NGCs' mean diameter was 0.2 ± 0.2 mm2 . PCR was positive for viral genomes in 16% of cases; NGCs were not statistically different in viral and non-viral myocarditis. WB analysis revealed a higher expression of HMGB1 in myocarditis compared to myocardial controls., Conclusions: NGCs are constantly recognizable in acute human myocarditis. Their number declines with age. Their integration within necrotic myocytes allows for the preservation of the cardiac structure and function.- Published
- 2024
- Full Text
- View/download PDF
17. Epicardial EMT and cardiac repair: an update.
- Author
-
Foglio E, D'Avorio E, Nieri R, Russo MA, and Limana F
- Subjects
- Humans, Animals, Heart Diseases metabolism, Heart Diseases pathology, Heart Diseases therapy, Myocardium metabolism, Myocardium pathology, Cell Differentiation, Epithelial-Mesenchymal Transition, Pericardium metabolism, Pericardium cytology
- Abstract
Epicardial epithelial-to-mesenchymal transition (EMT) plays a pivotal role in both heart development and injury response and involves dynamic cellular changes that are essential for cardiogenesis and myocardial repair. Specifically, epicardial EMT is a crucial process in which epicardial cells lose polarity, migrate into the myocardium, and differentiate into various cardiac cell types during development and repair. Importantly, following EMT, the epicardium becomes a source of paracrine factors that support cardiac growth at the last stages of cardiogenesis and contribute to cardiac remodeling after injury. As such, EMT seems to represent a fundamental step in cardiac repair. Nevertheless, endogenous EMT alone is insufficient to stimulate adequate repair. Redirecting and amplifying epicardial EMT pathways offers promising avenues for the development of innovative therapeutic strategies and treatment approaches for heart disease. In this review, we present a synthesis of recent literature highlighting the significance of epicardial EMT reactivation in adult heart disease patients., (© 2024. The Author(s).)
- Published
- 2024
- Full Text
- View/download PDF
18. Doxorubicin-Induced Cardiac Senescence Is Alleviated Following Treatment with Combined Polyphenols and Micronutrients through Enhancement in Mitophagy.
- Author
-
Foglio E, D'Avorio E, Vitiello L, Masuelli L, Bei R, Pacifici F, Della-Morte D, Mirabilii S, Ricciardi MR, Tafuri A, Garaci E, Russo MA, Tafani M, and Limana F
- Subjects
- Reactive Oxygen Species metabolism, Micronutrients, Cellular Senescence, Doxorubicin pharmacology, Mitophagy genetics, Sirtuin 3 genetics
- Abstract
Oxidative stress and impaired mitophagy are the hallmarks of cardiomyocyte senescence. Specifically, a decrease in mitophagic flux leads to the accumulation of damaged mitochondria and the development of senescence through increased ROS and other mediators. In this study, we describe the preventive role of A5
+ , a mix of polyphenols and other micronutrients, in doxorubicin (DOXO)-induced senescence of H9C2 cells. Specifically, H9C2 cells exposed to DOXO showed an increase in the protein expression proteins of senescence-associated genes, p21 and p16, and a decrease in the telomere binding factors TRF1 and TRF2, indicative of senescence induction. Nevertheless, A5+ pre-treatment attenuated the senescent-like cell phenotype, as evidenced by inhibition of all senescent markers and a decrease in SA-β-gal staining in DOXO-treated H9C2 cells. Importantly, A5+ restored the LC3 II/LC3 I ratio, Parkin and BNIP3 expression, therefore rescuing mitophagy, and decreased ROS production. Further, A5+ pre-treatment determined a ripolarization of the mitochondrial membrane and improved basal respiration. A5+ -mediated protective effects might be related to its ability to activate mitochondrial SIRT3 in synergy with other micronutrients, but in contrast with SIRT4 activation. Accordingly, SIRT4 knockdown in H9C2 cells further increased MnSOD activity, enhanced mitophagy, and reduced ROS generation following A5+ pre-treatment and DOXO exposure compared to WT cells. Indeed, we demonstrated that A5+ protects H9C2 cells from DOXO-induced senescence, establishing a new specific role for A5+ in controlling mitochondrial quality control by restoring SIRT3 activity and mitophagy, which provided a molecular basis for the development of therapeutic strategies against cardiomyocyte senescence.- Published
- 2023
- Full Text
- View/download PDF
19. The PTX3/TLR4 autocrine loop as a novel therapeutic target in triple negative breast cancer.
- Author
-
Giacomini A, Turati M, Grillo E, Rezzola S, Ghedini GC, Schuind AC, Foglio E, Maccarinelli F, Faletti J, Filiberti S, Chambery A, Valletta M, Melocchi L, Gofflot S, Chiavarina B, Turtoi A, Presta M, and Ronca R
- Abstract
Background: The pattern recognition receptor long pentraxin-3 (PTX3) plays conflicting roles in cancer by acting as an oncosuppressor or as a pro-tumor mediator depending on tumor context. Triple negative breast cancer (TNBC) represents the most aggressive histotype of breast cancer, characterized by the lack of efficacious therapeutic targets/approaches and poor prognosis. Thus, the characterization of new molecular pathways and/or alternative druggable targets is of great interest in TNBC., Methods: The expression of PTX3 in BC tumor samples and in BC cell lines has been analyzed using the Gene Expression-Based Outcome for Breast Cancer Online (GOBO), qPCR, Western blot and ELISA assay. The contribution of tumor and stromal cells to PTX3 production in TNBC was assessed by analyzing single cell RNA sequencing data and RNAscope performed on TNBC tumor samples. In order to investigate the effects of PTX3 in TNBC, different cell lines were engineered to knock-down (MDA-MB-231 and BT549 cells) or overexpress (MDA-MB-468 and E0771 cells) PTX3. Finally, using these engineered cells, in vitro (including gene expression profiling and gene set enrichment analyses) and in vivo (orthotopic tumor models in immune-compromised and immune competent mice) analyses were performed to assess the role and the molecular mechanism(s) exerted by PTX3 in TNBC., Results: In silico and experimental data indicate that PTX3 is mainly produced by tumor cells in TNBC and that its expression levels correlate with tumor stage. Accordingly, gene expression and in vitro results demonstrate that PTX3 overexpression confers a high aggressive/proliferative phenotype and fosters stem-like features in TNBC cells. Also, PTX3 expression induces a more tumorigenic potential when TNBC cells are grafted orthotopically in vivo. Conversely, PTX3 downregulation results in a less aggressive behavior of TNBC cells. Mechanistically, our data reveal that PTX3 drives the activation of the pro-tumorigenic Toll-like receptor 4 (TLR4) signaling pathway in TNBC, demonstrating for the first time that the PTX3/TLR4 autocrine stimulation loop contributes to TNBC aggressiveness and that TLR4 inhibition significantly impacts the growth of PTX3-producing TNBC cells., Conclusion: Altogether, these data shed light on the role of tumor-produced PTX3 in TNBC and uncover the importance of the PTX3/TLR4 axis for therapeutic and prognostic exploitation in TNBC., (© 2023. YUMED Inc. and BioMed Central Ltd.)
- Published
- 2023
- Full Text
- View/download PDF
20. The TFEB-TGIF1 axis regulates EMT in mouse epicardial cells.
- Author
-
Astanina E, Doronzo G, Corà D, Neri F, Oliviero S, Genova T, Mussano F, Middonti E, Vallariello E, Cencioni C, Valdembri D, Serini G, Limana F, Foglio E, Ballabio A, and Bussolino F
- Subjects
- Animals, Cells, Cultured, Mice, Organogenesis, Pericardium metabolism, Epithelial-Mesenchymal Transition physiology, Transforming Growth Factor beta metabolism
- Abstract
Epithelial-mesenchymal transition (EMT) is a complex and pivotal process involved in organogenesis and is related to several pathological processes, including cancer and fibrosis. During heart development, EMT mediates the conversion of epicardial cells into vascular smooth muscle cells and cardiac interstitial fibroblasts. Here, we show that the oncogenic transcription factor EB (TFEB) is a key regulator of EMT in epicardial cells and that its genetic overexpression in mouse epicardium is lethal due to heart defects linked to impaired EMT. TFEB specifically orchestrates the EMT-promoting function of transforming growth factor (TGF) β, and this effect results from activated transcription of thymine-guanine-interacting factor (TGIF)1, a TGFβ/Smad pathway repressor. The Tgif1 promoter is activated by TFEB, and in vitro and in vivo findings demonstrate its increased expression when Tfeb is overexpressed. Furthermore, Tfeb overexpression in vitro prevents TGFβ-induced EMT, and this effect is abolished by Tgif1 silencing. Tfeb loss of function, similar to that of Tgif1, sensitizes cells to TGFβ, inducing an EMT response to low doses of TGFβ. Together, our findings reveal an unexpected function of TFEB in regulating EMT, which might provide insights into injured heart repair and control of cancer progression., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
21. HMGB1-Mediated Activation of the Inflammatory-Reparative Response Following Myocardial Infarction.
- Author
-
Foglio E, Pellegrini L, Russo MA, and Limana F
- Subjects
- Alarmins metabolism, Animals, Humans, Oxidation-Reduction, Regeneration, HMGB1 Protein metabolism, Inflammation metabolism, Inflammation pathology, Myocardial Infarction metabolism, Myocardial Infarction pathology
- Abstract
Different cell types belonging to the innate and adaptive immune system play mutually non-exclusive roles during the different phases of the inflammatory-reparative response that occurs following myocardial infarction. A timely and finely regulation of their action is fundamental for the process to properly proceed. The high-mobility group box 1 (HMGB1), a highly conserved nuclear protein that in the extracellular space can act as a damage-associated molecular pattern (DAMP) involved in a large variety of different processes, such as inflammation, migration, invasion, proliferation, differentiation, and tissue regeneration, has recently emerged as a possible regulator of the activity of different immune cell types in the distinct phases of the inflammatory reparative process. Moreover, by activating endogenous stem cells, inducing endothelial cells, and by modulating cardiac fibroblast activity, HMGB1 could represent a master regulator of the inflammatory and reparative responses following MI. In this review, we will provide an overview of cellular effectors involved in these processes and how HMGB1 intervenes in regulating each of them. Moreover, we will summarize HMGB1 roles in regulating other cell types that are involved in the different phases of the inflammatory-reparative response, discussing how its redox status could affect its activity.
- Published
- 2022
- Full Text
- View/download PDF
22. miR-200c-3p Regulates Epitelial-to-Mesenchymal Transition in Epicardial Mesothelial Cells by Targeting Epicardial Follistatin-Related Protein 1.
- Author
-
Pontemezzo E, Foglio E, Vernucci E, Magenta A, D'Agostino M, Sileno S, Astanina E, Bussolino F, Pellegrini L, Germani A, Russo MA, and Limana F
- Subjects
- Animals, Biomarkers metabolism, Female, Mesoderm pathology, Mice, Inbred C57BL, MicroRNAs genetics, Neoplastic Stem Cells drug effects, Neoplastic Stem Cells metabolism, Neoplastic Stem Cells pathology, Transforming Growth Factor beta1 pharmacology, Mice, Epithelial-Mesenchymal Transition genetics, Epithelium metabolism, Follistatin-Related Proteins metabolism, MicroRNAs metabolism, Pericardium pathology
- Abstract
Recent findings suggest that epithelial to mesenchymal transition (EMT), a key step during heart development, is involved in cardiac tissue repair following myocardial infarction (MI). MicroRNAs (miRNAs) act as key regulators in EMT processes; however, the mechanisms by which miRNAs target epicardial EMT remain largely unknown. Here, by using an in vitro model of epicardial EMT, we investigated the role of miRNAs as regulators of this process and their potential targets. EMT was induced in murine epicardial-mesothelial cells (EMCs) through TGF β1 treatment for 48, 72, and 96 h as indicated by the expression of EMT-related genes by qRT-PCR, WB, and immunofluorescence. Further, enhanced expression of stemness genes was also detected. Among several EMT-related miRNAs, miR-200c-3p expression resulted as the most strongly suppressed. Interestingly, we also found a significant upregulation of Follistatin-related protein 1 (FSTL1), a miR-200c predicted target already identified as a potent cardiogenic factor produced by epicardial cells that promotes regeneration following MI. Dual-luciferase reporter assay demonstrated that miR-200c-3p directly targeted the 3'-untranslated region of FSTL1 in EMCs. Consistently, WB analysis showed that knockdown of miR-200c-3p significantly increased FSTL1 expression, whereas overexpression of miR-200c-3p counteracted TGF β1-mediated FSTL1 upregulation. Importantly, FSTL1 silencing maintained epithelial features in EMCs, despite EMT induction by TGF β1, and attenuated EMT-associated traits, including migration and stemness. In conclusion, epicardial FSTL1, an important cardiogenic factor in its secreted form, induces EMT, stemness, and migration of EMCs in a miR-200c-3p dependent pathway.
- Published
- 2021
- Full Text
- View/download PDF
23. FGF Trapping Inhibits Multiple Myeloma Growth through c-Myc Degradation-Induced Mitochondrial Oxidative Stress.
- Author
-
Ronca R, Ghedini GC, Maccarinelli F, Sacco A, Locatelli SL, Foglio E, Taranto S, Grillo E, Matarazzo S, Castelli R, Paganini G, Desantis V, Cattane N, Cattaneo A, Mor M, Carlo-Stella C, Belotti A, Roccaro AM, Presta M, and Giacomini A
- Subjects
- Animals, Apoptosis drug effects, Apoptosis physiology, Cell Line, Tumor, Cholesterol analogs & derivatives, Cholesterol pharmacology, Female, Fibroblast Growth Factors antagonists & inhibitors, Humans, Mice, Mice, Inbred NOD, Mice, SCID, Mitochondria drug effects, Mitochondria pathology, Multiple Myeloma drug therapy, Multiple Myeloma pathology, Random Allocation, Receptors, Fibroblast Growth Factor antagonists & inhibitors, Receptors, Fibroblast Growth Factor metabolism, Signal Transduction drug effects, Xenograft Model Antitumor Assays, Zebrafish, Fibroblast Growth Factors metabolism, Mitochondria metabolism, Multiple Myeloma metabolism, Oxidative Stress physiology, Proto-Oncogene Proteins c-myc metabolism
- Abstract
Multiple myeloma, the second most common hematologic malignancy, frequently relapses because of chemotherapeutic resistance. Fibroblast growth factors (FGF) act as proangiogenic and mitogenic cytokines in multiple myeloma. Here, we demonstrate that the autocrine FGF/FGFR axis is essential for multiple myeloma cell survival and progression by protecting multiple myeloma cells from oxidative stress-induced apoptosis. In keeping with the hypothesis that the intracellular redox status can be a target for cancer therapy, FGF/FGFR blockade by FGF trapping or tyrosine kinase inhibitor impaired the growth and dissemination of multiple myeloma cells by inducing mitochondrial oxidative stress, DNA damage, and apoptotic cell death that were prevented by the antioxidant vitamin E or mitochondrial catalase overexpression. In addition, mitochondrial oxidative stress occurred as a consequence of proteasomal degradation of the c-Myc oncoprotein that led to glutathione depletion. Accordingly, expression of a proteasome-nondegradable c-Myc protein mutant was sufficient to avoid glutathione depletion and rescue the proapoptotic effects due to FGF blockade. These findings were confirmed on bortezomib-resistant multiple myeloma cells as well as on bone marrow-derived primary multiple myeloma cells from newly diagnosed and relapsed/refractory patients, including plasma cells bearing the t(4;14) translocation obtained from patients with high-risk multiple myeloma. Altogether, these findings dissect the mechanism by which the FGF/FGFR system plays a nonredundant role in multiple myeloma cell survival and disease progression, and indicate that FGF targeting may represent a therapeutic approach for patients with multiple myeloma with poor prognosis and advanced disease stage. SIGNIFICANCE: This study provides new insights into the mechanisms by which FGF antagonists promote multiple myeloma cell death. GRAPHICAL ABSTRACT: http://cancerres.aacrjournals.org/content/canres/80/11/2340/F1.large.jpg., (©2020 American Association for Cancer Research.)
- Published
- 2020
- Full Text
- View/download PDF
24. MicroRNAs in Cancer Treatment-Induced Cardiotoxicity.
- Author
-
Pellegrini L, Sileno S, D'Agostino M, Foglio E, Florio MC, Guzzanti V, Russo MA, Limana F, and Magenta A
- Abstract
Cancer treatment has made significant progress in the cure of different types of tumors. Nevertheless, its clinical use is limited by unwanted cardiotoxicity. Aside from the conventional chemotherapy approaches, even the most newly developed, i.e., molecularly targeted therapy and immunotherapy, exhibit a similar frequency and severity of toxicities that range from subclinical ventricular dysfunction to severe cardiomyopathy and, ultimately, congestive heart failure. Specific mechanisms leading to cardiotoxicity still remain to be elucidated. For instance, oxidative stress and DNA damage are considered key players in mediating cardiotoxicity in different treatments. microRNAs (miRNAs) act as key regulators in cell proliferation, cell death, apoptosis, and cell differentiation. Their dysregulation has been associated with adverse cardiac remodeling and toxicity. This review provides an overview of the cardiotoxicity induced by different oncologic treatments and potential miRNAs involved in this effect that could be used as possible therapeutic targets.
- Published
- 2020
- Full Text
- View/download PDF
25. Cardiac Repair: The Intricate Crosstalk between the Epicardium and the Myocardium.
- Author
-
Pellegrini L, Foglio E, Pontemezzo E, Germani A, Russo MA, and Limana F
- Subjects
- Humans, Myocytes, Cardiac, Epithelial-Mesenchymal Transition, Myocardial Infarction, Myocardium, Pericardium physiology, Signal Transduction
- Abstract
Background: Substantial evidences support the hypothesis that the epicardium has a role in cardiac repair and regeneration in part providing, by epithelial to mesenchymal transition (EMT), progenitor cells that differentiate into cardiac cell types and in part releasing paracrine factors that contribute to cardiac repair. Besides cell contribution, a significant paracrine communication occurs between the epicardium and the myocardium that improves the whole regenerative response. Signaling pathways underlying this communication are multiple as well as soluble factors involved in cardiac repair and secreted both by myocardial and epicardial cells. Most recently, extracellular vesicles, i.e. exosomes, that accumulate in the pericardial fluid (PF) and are able to transport bioactive molecules (cytosolic proteins, mRNAs, miRNAs and other non-coding RNAs), have been also identified as potential mediators of epicardial-mediated repair following myocardial injury., Conclusion: This mini-review provides an overview of the epicardial-myocardial signaling in regulating cardiac repair in ischemic heart diseases. Indeed, a detailed understanding of the crosstalk between myocardial and epicardial cells and how paracrine mechanisms are involved in the context of ischemic heart diseases would be of tremendous help in developing novel therapeutic approaches to promote cardiomyocytes survival and heart regeneration following myocardial infarction (MI)., (Copyright© Bentham Science Publishers; For any queries, please email at epub@benthamscience.net.)
- Published
- 2020
- Full Text
- View/download PDF
26. HMGB1-mediated apoptosis and autophagy in ischemic heart diseases.
- Author
-
Foglio E, Pellegrini L, Germani A, Russo MA, and Limana F
- Abstract
Acute myocardial infarction (MI) and its consequences are the most common and lethal heart syndromes worldwide and represent a significant health problem. Following MI, apoptosis has been generally seen as the major contributor of the cardiomyocyte fate and of the resultant myocardial remodeling. However, in recent years, it has been discovered that, following MI, cardiomyocytes could activate autophagy in an attempt to protect themselves against ischemic stress and to preserve cardiac function. Although initially seen as two completely separate responses, recent works have highlighted the intertwined crosstalk between apoptosis and autophagy. Numerous researches have tried to unveil the mechanisms and the molecular players involved in this phenomenon and have identified in high-mobility group box 1 (HMGB1), a highly conserved non-histone nuclear protein with important roles in the heart, one of the major regulator. Thus, the aim of this mini review is to discuss how HMGB1 regulates these two responses in ischemic heart diseases. Indeed, a detailed understanding of the crosstalk between apoptosis and autophagy in these pathologies and how HMGB1 regulates them would be of tremendous help in developing novel therapeutic approaches aimed to promote cardiomyocyte survival and to diminish tissue injury following MI., Competing Interests: The authors declare that there is no conflict of interest that could be perceived as prejudicing the impartiality of this review., (© 2019 The authors.)
- Published
- 2019
- Full Text
- View/download PDF
27. HMGB1 and repair: focus on the heart.
- Author
-
Pellegrini L, Foglio E, Pontemezzo E, Germani A, Russo MA, and Limana F
- Subjects
- Animals, Heart Diseases pathology, Humans, Myocardium metabolism, Myocardium pathology, HMGB1 Protein metabolism, Heart Diseases metabolism
- Abstract
High-mobility group box 1 (HMGB1) is one of the most abundant proteins in eukaryotes and the best characterized damage-associated molecular pattern (DAMP). The biological activities of HMGB1 depend on its subcellular location, context and post-translational modifications. Inside the nucleus, HMGB1 is engaged in many DNA events such as DNA repair, transcription regulation and genome stability; in the cytoplasm, its main function is to regulate the autophagic flux while in the extracellular environment, it possesses more complicated functions and it is involved in a large variety of different processes such as inflammation, migration, invasion, proliferation, differentiation and tissue regeneration. Due to this pleiotropy, the role of HMGB1 has been vastly investigated in various pathological diseases and a large number of studies have explored its function in cardiovascular pathologies. However, in this contest, the precise mechanism of action of HMGB1 and its therapeutic potential are still very controversial since is debated whether HMGB1 is involved in tissue damage or plays a role in tissue repair and regeneration. The main focus of this review is to provide an overview of the effects of HMGB1 in different ischemic heart diseases and to discuss its functions in these pathological conditions., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
28. Long Pentraxin 3-Mediated Fibroblast Growth Factor Trapping Impairs Fibrosarcoma Growth.
- Author
-
Rodrigues PF, Matarazzo S, Maccarinelli F, Foglio E, Giacomini A, Silva Nunes JP, Presta M, Dias AAM, and Ronca R
- Abstract
Fibrosarcomas are soft tissue mesenchymal tumors originating from transformed fibroblasts. Fibroblast growth factor-2 (FGF2) and its tyrosine-kinase receptors (FGFRs) play pivotal roles in fibrosarcoma onset and progression, FGF2 being actively produced by fibroblasts in all stages along their malignant transformation to the fibrosarcoma stage. The soluble pattern recognition receptor long pentraxin-3 (PTX3) is an extrinsic oncosuppressor whose expression is reduced in different tumor types, including soft tissue sarcomas, via hypermethylation of its gene promoter. PTX3 interacts with FGF2 and other FGF family members, thus acting as a multi-FGF antagonist able to inhibit FGF-dependent neovascularization and tumor growth. Here, PTX3 overexpression significantly reduced the proliferative and tumorigenic potential of fibrosarcoma cells in vitro and in vivo . In addition, systemic delivery of human PTX3 driven by the Tie2 promoter inhibited the growth of fibrosarcoma grafts in transgenic mice. In a translational perspective, the PTX3-derived small molecule FGF trap NSC12 prevented activation of the FGF/FGFR system in fibrosarcoma cells and reduced their tumorigenic activity in vivo . In conclusion, impairment of the FGF/FGFR system by FGF trap molecules may represent a novel therapeutic approach for the treatment of fibrosarcoma.
- Published
- 2018
- Full Text
- View/download PDF
29. Long Pentraxin-3 Modulates the Angiogenic Activity of Fibroblast Growth Factor-2.
- Author
-
Presta M, Foglio E, Churruca Schuind A, and Ronca R
- Subjects
- Animals, Humans, Inflammation, Neovascularization, Physiologic, C-Reactive Protein physiology, Fibroblast Growth Factor 2 metabolism, Serum Amyloid P-Component physiology
- Abstract
Angiogenesis, the process of new blood vessel formation from pre-existing ones, plays a key role in various physiological and pathological conditions. Alteration of the angiogenic balance, consequent to the deranged production of angiogenic growth factors and/or natural angiogenic inhibitors, is responsible for angiogenesis-dependent diseases, including cancer. Fibroblast growth factor-2 (FGF2) represents the prototypic member of the FGF family, able to induce a complex "angiogenic phenotype" in endothelial cells in vitro and a potent neovascular response in vivo as the consequence of a tight cross talk between pro-inflammatory and angiogenic signals. The soluble pattern recognition receptor long pentraxin-3 (PTX3) is a member of the pentraxin family produced locally in response to inflammatory stimuli. Besides binding features related to its role in innate immunity, PTX3 interacts with FGF2 and other members of the FGF family via its N-terminal extension, thus inhibiting FGF-mediated angiogenic responses in vitro and in vivo . Accordingly, PTX3 inhibits the growth and vascularization of FGF-dependent tumors and FGF2-mediated smooth muscle cell proliferation and artery restenosis. Recently, the characterization of the molecular bases of FGF2/PTX3 interaction has allowed the identification of NSC12, the first low molecular weight pan-FGF trap able to inhibit FGF-dependent tumor growth and neovascularization. The aim of this review is to provide an overview of the impact of PTX3 and PTX3-derived molecules on the angiogenic, inflammatory, and tumorigenic activity of FGF2 and their potential implications for the development of more efficacious anti-FGF therapeutic agents to be used in those clinical settings in which FGFs play a pathogenic role.
- Published
- 2018
- Full Text
- View/download PDF
30. Molecular mechanisms of cardioprotective effects mediated by transplanted cardiac ckit + cells through the activation of an inflammatory hypoxia-dependent reparative response.
- Author
-
Puddighinu G, D'Amario D, Foglio E, Manchi M, Siracusano A, Pontemezzo E, Cordella M, Facchiano F, Pellegrini L, Mangoni A, Tafani M, Crea F, Germani A, Russo MA, and Limana F
- Abstract
The regenerative effects of cardiac ckit
+ stem cells (ckit+ CSCs) in acute myocardial infarction (MI) have been studied extensively, but how these cells exert a protective effect on cardiomyocytes is not well known. Growing evidences suggest that in adult stem cells injury triggers inflammatory signaling pathways which control tissue repair and regeneration. Aim of the present study was to determine the mechanisms underlying the cardioprotective effects of ckit+ CSCs following transplantation in a murine model of MI. Following isolation and in vitro expansion, cardiac ckit+ CSCs were subjected to normoxic and hypoxic conditions and assessed at different time points. These cells adapted to hypoxia as showed by the activation of HIF-1α and the expression of a number of genes, such as VEGF, GLUT1, EPO, HKII and, importantly, of alarmin receptors, such as RAGE, P2X7R, TLR2 and TLR4. Activation of these receptors determined an NFkB-dependent inflammatory and reparative gene response (IRR). Importantly, hypoxic ckit+ CSCs increased the secretion of the survival growth factors IGF-1 and HGF. To verify whether activation of the IRR in a hypoxic microenvironment could exert a beneficial effect in vivo , autologous ckit+ CSCs were transplanted into mouse heart following MI. Interestingly, transplantation of ckit+ CSCs lowered apoptotic rates and induced autophagy in the peri-infarct area; further, it reduced hypertrophy and fibrosis and, most importantly, improved cardiac function. ckit+ CSCs are able to adapt to a hypoxic environment and activate an inflammatory and reparative response that could account, at least in part, for a protective effect on stressed cardiomyocytes following transplantation in the infarcted heart., Competing Interests: CONFLICTS OF INTEREST None declared.- Published
- 2017
- Full Text
- View/download PDF
31. SIRT1-SIRT3 Axis Regulates Cellular Response to Oxidative Stress and Etoposide.
- Author
-
Carnevale I, Pellegrini L, D'Aquila P, Saladini S, Lococo E, Polletta L, Vernucci E, Foglio E, Coppola S, Sansone L, Passarino G, Bellizzi D, Russo MA, Fini M, and Tafani M
- Subjects
- Acetylation drug effects, Animals, Cell Line, Tumor, Cytoprotection drug effects, Gene Silencing drug effects, HEK293 Cells, Humans, Intracellular Space metabolism, Mice, Models, Biological, Promoter Regions, Genetic genetics, Protein Binding drug effects, Reactive Oxygen Species metabolism, Sp1 Transcription Factor metabolism, Etoposide pharmacology, Oxidative Stress drug effects, Signal Transduction drug effects, Sirtuin 1 metabolism, Sirtuin 3 metabolism
- Abstract
Sirtuins are conserved NAD
+ -dependent deacylases. SIRT1 is a nuclear and cytoplasmic sirtuin involved in the control of histones a transcription factors function. SIRT3 is a mitochondrial protein, which regulates mitochondrial function. Although, both SIRT1 and SIRT3 have been implicated in resistance to cellular stress, the link between these two sirtuins has not been studied so far. Here we aimed to unravel: i) the role of SIRT1-SIRT3 axis for cellular response to oxidative stress and DNA damage; ii) how mammalian cells modulate such SIRT1-SIRT3 axis and which mechanisms are involved. Therefore, we analyzed the response to different stress stimuli in WT or SIRT1-silenced cell lines. Our results demonstrate that SIRT1-silenced cells are more resistant to H2 O2 and etoposide treatment showing decreased ROS accumulation, γ-H2AX phosphorylation, caspase-3 activation and PARP cleavage. Interestingly, we observed that SIRT1-silenced cells show an increased SIRT3 expression. To explore such a connection, we carried out luciferase assays on SIRT3 promoter demonstrating that SIRT1-silencing increases SIRT3 promoter activity and that such an effect depends on the presence of SP1 and ZF5 recognition sequences on SIRT3 promoter. Afterwards, we performed co-immunoprecipitation assays demonstrating that SIRT1 binds and deacetylates the transcription inhibitor ZF5 and that there is a decreased interaction between SP1 and ZF5 in SIRT1-silenced cells. Therefore, we speculate that acetylated ZF5 cannot bind and sequester SP1 that is free, then, to increase SIRT3 transcription. In conclusion, we demonstrate that cells with low SIRT1 levels can maintain their resistance and survival by increasing SIRT3 expression. J. Cell. Physiol. 232: 1835-1844, 2017. © 2016 Wiley Periodicals, Inc., (© 2016 Wiley Periodicals, Inc.)- Published
- 2017
- Full Text
- View/download PDF
32. HMGB1 Inhibits Apoptosis Following MI and Induces Autophagy via mTORC1 Inhibition.
- Author
-
Foglio E, Puddighinu G, Germani A, Russo MA, and Limana F
- Subjects
- AMP-Activated Protein Kinases metabolism, Animals, Biomarkers metabolism, Cell Separation, Cell Survival drug effects, Enzyme Activation drug effects, Female, Heart Function Tests, Mechanistic Target of Rapamycin Complex 1, Mice, Inbred C57BL, Multiprotein Complexes metabolism, Myocardial Infarction physiopathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Myocytes, Cardiac pathology, Signal Transduction drug effects, TOR Serine-Threonine Kinases metabolism, Ventricular Function, Left drug effects, Ventricular Remodeling drug effects, Apoptosis drug effects, Autophagy drug effects, HMGB1 Protein pharmacology, Multiprotein Complexes antagonists & inhibitors, Myocardial Infarction pathology, TOR Serine-Threonine Kinases antagonists & inhibitors
- Abstract
Exogenous High Mobility Group Box-1 protein (HMGB1) has been reported to protect the infarcted heart but the underlying mechanism is quite complex. In particular, its effect on ischemic cardiomyocytes has been poorly investigated. Aim of the present study was to verify whether and how autophagy and apoptosis were involved in HMGB1-induced heart repair following myocardial infarction (MI). HMGB1 (200 ng) or denatured HMGB1 were injected in the peri-infarcted region of mouse hearts following acute MI. Three days after treatment, an upregulation of autophagy was detected in infarcted HMGB1 treated hearts compared to controls. Specifically, HMGB1 induced autophagy by significantly upregulating the protein expression of LC3, Beclin-1, and Atg7 in the border zone. To gain further insights into the molecular mechanism of HMGB1-mediated autophagy, WB analysis were performed in cardiomyocytes isolated from 3 days infarcted hearts in the presence and in the absence of HMGB1 treatment. Results showed that upregulation of autophagy by HMGB1 treatment was potentially related to activation of AMP-activated protein kinase (AMPK) and inhibition of the mammalian target of rapamycin complex 1 (mTORC1). Accordingly, in these hearts, phospho-Akt signaling pathway was inhibited. The induction of autophagy was accompanied by reduced cardiomyocyte apoptotic rate and decreased expression levels of Bax/Bcl-2 and active caspase-3 in the border zone of 3 days infarcted mice following HMGB1 treatment. We report the first in vivo evidence that HMGB1 treatment in a murine model of acute MI might induce cardiomyocyte survival through attenuation of apoptosis and AMP-activated protein kinase-dependent autophagy. J. Cell. Physiol. 232: 1135-1143, 2017. © 2016 Wiley Periodicals, Inc., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
33. Exosomal clusterin, identified in the pericardial fluid, improves myocardial performance following MI through epicardial activation, enhanced arteriogenesis and reduced apoptosis.
- Author
-
Foglio E, Puddighinu G, Fasanaro P, D'Arcangelo D, Perrone GA, Mocini D, Campanella C, Coppola L, Logozzi M, Azzarito T, Marzoli F, Fais S, Pieroni L, Marzano V, Germani A, Capogrossi MC, Russo MA, and Limana F
- Subjects
- Aged, Aged, 80 and over, Animals, Apoptosis physiology, Biomarkers analysis, Biomarkers metabolism, Clusterin analysis, Coronary Vessels chemistry, Exosomes chemistry, Female, Humans, Male, Mice, Mice, Inbred C57BL, Middle Aged, Myocardial Infarction diagnosis, Myocardium chemistry, Myocardium metabolism, Myocardium pathology, Pericardial Fluid chemistry, Pericardium chemistry, Pericardium pathology, Clusterin metabolism, Coronary Vessels metabolism, Exosomes metabolism, Myocardial Infarction metabolism, Pericardial Fluid metabolism, Pericardium metabolism
- Abstract
Background: We recently demonstrated that epicardial progenitor cells participate in the regenerative response to myocardial infarction (MI) and factors released in the pericardial fluid (PF) may play a key role in this process. Exosomes are secreted nanovesicles of endocytic origin, identified in most body fluids, which may contain molecules able to modulate a variety of cell functions. Here, we investigated whether exosomes are present in the PF and their potential role in cardiac repair., Methods and Results: Early gene expression studies in 3day-infarcted mouse hearts showed that PF induces epithelial-to-mesenchymal transition (EMT) in epicardial cells. Exosomes were identified in PFs from non-infarcted patients (PFC) and patients with acute MI (PFMI). A shotgun proteomics analysis identified clusterin in exosomes isolated from PFMI but not from PFC. Notably, clusterin has a protective effect on cardiomyocytes after acute MI in vivo and is an important mediator of TGFβ-induced. Clusterin addition to the pericardial sac determined an increase in epicardial cells expressing the EMT marker α-SMA and, interestingly, an increase in the number of epicardial cells ckit(+)/α-SMA(+), 7days following MI. Importantly, clusterin treatment enhanced arteriolar length density and lowered apoptotic rates in the peri-infarct area. Hemodynamic studies demonstrated an improvement in cardiac function in clusterin-treated compared to untreated infarcted hearts., Conclusions: Exosomes are present and detectable in the PFs. Clusterin was identified in PFMI-exosomes and might account for an improvement in myocardial performance following MI through a framework including EMT-mediated epicardial activation, arteriogenesis and reduced cardiomyocyte apoptosis., (Copyright © 2015 Elsevier Ireland Ltd. All rights reserved.)
- Published
- 2015
- Full Text
- View/download PDF
34. Generation of cardiac progenitor cells through epicardial to mesenchymal transition.
- Author
-
Germani A, Foglio E, Capogrossi MC, Russo MA, and Limana F
- Subjects
- Cell Differentiation, Humans, Myocytes, Cardiac cytology, Signal Transduction, Epithelial-Mesenchymal Transition physiology, Mesenchymal Stem Cells cytology, Myocardial Ischemia pathology, Myocardium cytology, Pericardium cytology
- Abstract
The epithelial to mesenchymal transition (EMT) is a biological process that drives the formation of cells involved both in tissue repair and in pathological conditions, including tissue fibrosis and tumor metastasis by providing cancer cells with stem cell properties. Recent findings suggest that EMT is reactivated in the heart following ischemic injury. Specifically, epicardial EMT might be involved in the formation of cardiac progenitor cells (CPCs) that can differentiate into endothelial cells, smooth muscle cells, and, possibly, cardiomyocytes. The identification of mechanisms and signaling pathways governing EMT-derived CPC generation and differentiation may contribute to the development of a more efficient regenerative approach for adult heart repair. Here, we summarize key literature in the field.
- Published
- 2015
- Full Text
- View/download PDF
35. Transcriptional profiling of HMGB1-induced myocardial repair identifies a key role for Notch signaling.
- Author
-
Limana F, Esposito G, Fasanaro P, Foglio E, Arcelli D, Voellenkle C, Di Carlo A, Avitabile D, Martelli F, Russo MA, Pompilio G, Germani A, and C Capogrossi M
- Subjects
- Animals, Cell Differentiation drug effects, Cell Proliferation drug effects, Cells, Cultured, Female, HMGB1 Protein administration & dosage, Heart drug effects, Mice, Mice, Inbred C57BL, Myocardial Infarction physiopathology, Myocytes, Cardiac drug effects, Myocytes, Cardiac metabolism, Regeneration drug effects, Gene Expression Profiling, HMGB1 Protein pharmacology, Myocardial Infarction metabolism, Myocardium metabolism, Receptors, Notch metabolism, Regeneration genetics, Signal Transduction drug effects
- Abstract
Exogenous high-mobility group box 1 protein (HMGB1) administration to the mouse heart, during acute myocardial infarction (MI), results in cardiac regeneration via resident c-kit(+) cell (CPC) activation. Aim of the present study was to identify the molecular pathways involved in HMGB1-induced heart repair. Gene expression profiling was performed to identify differentially expressed genes in the infarcted and bordering regions of untreated and HMGB1-treated mouse hearts, 3 days after MI. Functional categorization of the transcripts, accomplished using Ingenuity Pathway Analysis software (IPA), revealed that genes involved in tissue regeneration, that is, cardiogenesis, vasculogenesis and angiogenesis, were present both in the infarcted area and in the peri-infarct zone; HMGB1 treatment further increased the expression of these genes. IPA revealed the involvement of Notch signaling pathways in HMGB1-treated hearts. Importantly, HMGB1 determined a 35 and 58% increase in cardiomyocytes and CPCs expressing Notch intracellular cytoplasmic domain, respectively. Further, Notch inhibition by systemic treatment with the γ-secretase inhibitor DAPT, which blocked the proteolytic activation of Notch receptors, reduced the number of CPCs, their proliferative fraction, and cardiomyogenic differentiation in HMGB1-treated infarcted hearts. The present study gives insight into the molecular processes involved in HMGB1-mediated cardiac regeneration and indicates Notch signaling as a key player.
- Published
- 2013
- Full Text
- View/download PDF
36. Silicic acid in drinking water prevents age-related alterations in the endothelium-dependent vascular relaxation modulating eNOS and AQP1 expression in experimental mice: an immunohistochemical study.
- Author
-
Buffoli B, Foglio E, Borsani E, Exley C, Rezzani R, and Rodella LF
- Subjects
- Administration, Oral, Aging, Animals, Aorta, Thoracic pathology, Disease Models, Animal, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Immunohistochemistry, Kidney blood supply, Kidney pathology, Male, Mice, Mice, Inbred C57BL, Vasodilation physiology, Aorta, Thoracic metabolism, Aquaporin 1 metabolism, Drinking Water administration & dosage, Endothelium, Vascular drug effects, Nitric Oxide Synthase Type III metabolism, Silicic Acid administration & dosage, Vasodilation drug effects
- Abstract
The maintenance of endothelial integrity is of great importance in coping with age-related vascular alterations. Endothelium-derived nitric oxide is one of the various vasoactive substances able to regulate vascular tone and homeostasis, and whose decrease is known to be related with senescence in endothelial cells. There are reports on the efficacy of silicon, especially as silicic acid, in protecting vascular integrity during age-related vascular diseases. The aim of this study was to evaluate the ability of supplementation of silicic acid in drinking water in the maintenance of vascular health in a mouse model of early physiological aging. In particular, we evaluated the relationship between Si supplementation and endothelial nitric oxide synthase (eNOS) expression, taking into account also the aquaporin-1 (AQP-1) isoform that, as recently reported, seems to be involved in nitric oxide transport across cell membranes. Our results showed that silicic acid supplementation increased both eNOS and AQP-1 expression, suggesting that silicic acid modulation of endothelial nitric oxide synthase and aquaporin-1 could represent a potential strategy against age-related vascular senescence., (Copyright © 2012 Elsevier GmbH. All rights reserved.)
- Published
- 2013
- Full Text
- View/download PDF
37. Aging and vascular dysfunction: beneficial melatonin effects.
- Author
-
Rodella LF, Favero G, Rossini C, Foglio E, Bonomini F, Reiter RJ, and Rezzani R
- Subjects
- Animals, Antioxidants pharmacology, Atherosclerosis pathology, Atherosclerosis physiopathology, Cells, Cultured, Disease Models, Animal, Endothelium, Vascular metabolism, Endothelium, Vascular pathology, Male, Mice, Mice, Inbred C57BL, Radioimmunoassay, Up-Regulation drug effects, Aging physiology, Atherosclerosis prevention & control, Endothelium, Vascular physiopathology, Melatonin pharmacology, Oxidative Stress, Vasodilation drug effects
- Abstract
Aging is characterized by a progressive deterioration of physiological functions and metabolic processes. In aging and in diseases associated with the elderly, the loss of cells in vital structures or organs may be related to several factors. Sirtuin1 (SIRT1) is a member of the sirtuin family of protein deacetylases involved in life span extension; however, its involvement in the aging is not yet completely defined. Recently, melatonin, a pleiotropic molecule, shown to activate SIRT1 in primary neurons of young animals, as well as in aged neurons of a murine model of senescence. Melatonin is known to modulate oxidative stress-induced senescence and pro-survival pathways. We treated 6- and 15-week-old apolipoprotein E (APOE)-deficient mice (APOE 6w and 15w) with two melatonin formulations (FAST and RETARD) to evaluate their anti-aging effect. Morphological changes in vessels (aortic arch) of APOE mice were evaluated SIRT1, p53, endothelial nitric oxide synthase (eNOS), and endothelin-1 (ET-1) markers. We demonstrate that SIRT1 and eNOS decresed in APOE mice between 6 and 15 weeks and that aging induced an elevated expression of p53 and ET-1 in APOE animals. Melatonin improved the impairment of endothelial damage and reduced loss of SIRT1 and eNOS decreasing p53 and ET-1 expression. The RETARD melatonin preparation caused a greater improvement of vessel cytoarchitecture. In summary, we indicate that SIRT1-p53-eNOS axis as one of the important marker of advanced vascular dysfunctions linked to aging. Finally, we suggest that extended-release melatonin (RETARD) provides a more appropriate option for contrasting these dysfunctions compared with rapid release melatonin (FAST) administration.
- Published
- 2013
- Full Text
- View/download PDF
38. Vascular endothelial cells and dysfunctions: role of melatonin.
- Author
-
Rodella LF, Favero G, Foglio E, Rossini C, Castrezzati S, Lonati C, and Rezzani R
- Subjects
- Atherosclerosis metabolism, Diabetes Mellitus metabolism, Humans, Nicotine toxicity, Vascular Diseases chemically induced, Atherosclerosis physiopathology, Diabetes Mellitus physiopathology, Endothelial Cells metabolism, Hypertension metabolism, Melatonin metabolism, Reperfusion Injury metabolism, Vascular Diseases physiopathology
- Abstract
Several pathological conditions, including hypertension, atherosclerosis, diabetes, ischemia/reperfusion injury and nicotine-induced vasculopathy, are associated with vascular endothelial dysfunction characterized by altered secretory output of endothelial cells. Therefore there is a search for molecules and interventions that could restore endothelial function, in particular augmenting NO production, reducing the generation of free radicals and vasoconstrictors and preventing undesired inflammation. The pineal hormone melatonin exhibits several endothelium protective properties: it scavenges free radicals, activates antioxidant defence enzymes, normalizes lipid and blood pressure profile and increases NO bioavailability. Melatonin improved vascular function in experimental hypertension, reducing intimal infiltration and restoring NO production. Melatonin improved the NO pathway also in animal models for the study of diabetes and prevented NO down-regulation and adhesive molecules up-regulation in nicotine-induced vasculopathy. The protection against endothelial damage, vasoconstriction, platelet aggregation and leukocyte infiltration might contribute to the beneficial effects against ischemia-reperfusion injury by melatonin. Therefore, melatonin administration has endothelium-protective potential in several pathological conditions. Nevertheless, it still needs to be established, whether melatonin is able to revert already established endothelial dysfunction in these conditions.
- Published
- 2013
- Full Text
- View/download PDF
39. Regular consumption of a silicic acid-rich water prevents aluminium-induced alterations of nitrergic neurons in mouse brain: histochemical and immunohistochemical studies.
- Author
-
Foglio E, Buffoli B, Exley C, Rezzani R, and Rodella LF
- Subjects
- Alum Compounds analysis, Animals, Cerebral Cortex metabolism, Cerebral Cortex pathology, Drinking, Drinking Water chemistry, Drug Antagonism, Male, Mice, Mice, Inbred C57BL, Mineral Waters analysis, NADPH Dehydrogenase metabolism, Nitrergic Neurons metabolism, Nitrergic Neurons pathology, Nitric Oxide Synthase Type I metabolism, Toxicity Tests, Chronic, Water Pollutants, Chemical analysis, Alum Compounds toxicity, Cerebral Cortex drug effects, Neuroprotective Agents pharmacology, Nitrergic Neurons drug effects, Silicic Acid pharmacology, Water Pollutants, Chemical toxicity
- Abstract
Silicon is not generally considered an essential nutrient for mammals and, to date, whether it has a biological role or beneficial effects in humans is not known. The results of a number of studies suggest that dietary silicon supplementation might have a protective effect both for limiting aluminium absorption across the gut and for the removal of systemic aluminium via the urine, hence, preventing potential accumulation of aluminium in the brain. Since our previous studies demonstrated that aluminium exposure reduces the number of nitrergic neurons, the aim of the present study was to compare the distribution and the morphology of NO-containing neurons in brain cortex of mice exposed to aluminium sulphate dissolved in silicic acid-rich or poor drinking water to assess the potential protective role of silicon against aluminium toxicity in the brain. NADPH-d histochemistry and nNOS immunohistochemistry showed that high concentrations of silicon in drinking water were able to minimize the impairment of the function of nitrergic neurons induced by aluminium administration. We found that silicon protected against aluminium-induced damage to the nitrergic system: in particular, we demonstrated that silicon maintains the number of nitrergic neurons and their expression of nitrergic enzymes at physiological levels, even after a 12 and 15 month exposure to aluminium.
- Published
- 2012
- Full Text
- View/download PDF
40. Heme induced oxidative stress attenuates sirtuin1 and enhances adipogenesis in mesenchymal stem cells and mouse pre-adipocytes.
- Author
-
Puri N, Sodhi K, Haarstad M, Kim DH, Bohinc S, Foglio E, Favero G, and Abraham NG
- Subjects
- 3T3-L1 Cells, Adipocytes cytology, Adipocytes drug effects, Adiponectin biosynthesis, Animals, Antioxidants pharmacology, CCAAT-Enhancer-Binding Protein-alpha biosynthesis, Cell Line, Cyclic N-Oxides pharmacology, Fatty Acid-Binding Proteins biosynthesis, Humans, Mesenchymal Stem Cells cytology, Mesenchymal Stem Cells drug effects, Mice, PPAR gamma biosynthesis, Reactive Oxygen Species metabolism, Signal Transduction, Spin Labels, Adipocytes physiology, Adipogenesis drug effects, Heme metabolism, Heme pharmacology, Mesenchymal Stem Cells physiology, Oxidative Stress, Sirtuin 1 biosynthesis
- Abstract
Patho-physiological conditions with high oxidative stress, such as conditions associated with increased denatured heme-proteins, are associated with enhanced adipogenic response. This effect predominantly manifests as adipocyte hypertrophy characterized by dysfunctional, pro-inflammatory adipocytes exhibiting reduced expression of anti-inflammatory hormone, adiponectin. To understand how increased levels of cellular heme, a pro-oxidant molecule, modulates adipogenesis; the following study was designed to evaluate effects of heme on adipogenesis in human mesenchymal stem cells (hMSCs) and mouse pre-adipocytes (3T3L1). Experiments were conducted in the absence and in the presence of a superoxide dismutase (SOD) mimetic (tempol, 100 µM). Heme (10 µM) increased (P<0.05) adipogenesis in hMSCs and mouse pre-adipocytes, where tempol alone (100 µmol/L) attenuated adipogenesis in these cells (P<0.05). Tempol also reversed heme-induced increase in adipogenesis in both hMSCs and mouse pre-adipocytes (P<0.05). In addition, heme exposed 3T3L1 exhibited reduced (P<0.05) expression of transcriptional regulator-sirtuin 1 (Sirt1), along with, increased (P<0.05) expression of adipogenic markers peroxisome proliferators-activated receptor-gamma (PPARγ), C/EBPα, and aP2. These effects of heme were rescued (P<0.05) in cells concurrently treated with heme and tempol (P<0.05) and prevented in cells over-expressing Sirt1. Taken together, our results indicate that heme-induced oxidative stress inhibits Sirt1, thus un-inhibiting adipogenic regulators such as PPARγ and C/EBPα; which in turn induce increased adipogenesis along with adipocyte hypertrophy in pre-adipocytes. Anti-oxidant induced offsetting of these effects of heme supports the role of heme-dependent oxidative stress in mediating such events., (Copyright © 2012 Wiley Periodicals, Inc.)
- Published
- 2012
- Full Text
- View/download PDF
41. Nicotine-induced morphological changes in rat aorta: the protective role of melatonin.
- Author
-
Rodella LF, Rossini C, Favero G, Foglio E, Loreto C, and Rezzani R
- Subjects
- Animals, Aorta cytology, Aorta drug effects, Aorta metabolism, Cells, Cultured, Immunohistochemistry, Male, Muscle, Smooth, Vascular cytology, Muscle, Smooth, Vascular metabolism, Rats, Rats, Wistar, Melatonin pharmacology, Muscle, Smooth, Vascular drug effects, Nicotine pharmacology
- Abstract
We analyzed the morphological changes in rat aortas during nicotine administration in order to investigate the involvement of vascular smooth muscle cells (VSMCs) in the regulation of vascular wall homeostasis. We also considered the possibility of restoring VSMC changes using melatonin as an antioxidant. We studied 4 groups of animals over 56 days. Three groups of rats were used as controls (the first without treatment, the second with melatonin alone and the third with nicotine alone). The last group of rats was orally treated with nicotine for the first 28 days and with melatonin for the last 28 days. Morphological changes in vessels were evaluated by histological procedures and immunohistochemical analysis using thrombospondin-1 (TSP-1), transforming growth factor-β1 (TGF-β1), plasminogen activator inhibitor-1 (PAI-1) and CD31 antibodies. We demonstrated that TSP-1, TGF-β1 and PAI-1 increased after nicotine administration. We believe that TSP-1 is responsible for neointima formation and that it is able to influence TGF-β1 and PAI-1 expression. Histological and immunohistochemical analysis by CD31 antibody showed that only a few endothelial cells were present in the aorta after nicotine administration compared to controls and rats treated with melatonin after nicotine administration. Moreover, histological analysis showed that neointima formation was present after nicotine treatment. Furthermore, melatonin inhibited neointima formation increasing TSP-1 expression. The ability of melatonin to inhibit neointima formation suggests that it could be a useful treatment for homeostasis of vascular walls., (Copyright © 2011 S. Karger AG, Basel.)
- Published
- 2012
- Full Text
- View/download PDF
42. Endothelin-1 as a potential marker of melatonin's therapeutic effects in smoking-induced vasculopathy.
- Author
-
Rodella LF, Favero G, Rossini C, Foglio E, Reiter RJ, and Rezzani R
- Subjects
- Animals, Aorta drug effects, Aorta metabolism, Aorta physiopathology, Biomarkers metabolism, Cardiovascular Diseases physiopathology, Endothelium, Vascular drug effects, Endothelium, Vascular metabolism, Endothelium, Vascular physiopathology, HSP70 Heat-Shock Proteins biosynthesis, Male, Melatonin pharmacology, Nitric Oxide Synthase Type II biosynthesis, Nitric Oxide Synthase Type III biosynthesis, Random Allocation, Rats, Rats, Wistar, Smoking adverse effects, Up-Regulation physiology, Vasoconstriction drug effects, Vasoconstriction physiology, Vasodilation drug effects, Vasodilation physiology, Cardiovascular Diseases etiology, Cardiovascular Diseases metabolism, Endothelin-1 biosynthesis, Melatonin therapeutic use, Nicotine toxicity, Smoking metabolism
- Abstract
Aims: Smoking is a significant independent risk factor for cardiovascular disease. Among the chemicals present in the cigarette smoke, nicotine is responsible for much of the damage; it induces marked vessel morphological dysfunction and vasoconstriction. Unfortunately, pharmacological or behavioural treatment is not useful against cigarette smoking. The purpose of this study is to test, in experimental conditions, the therapeutic ability of exogenous melatonin administered after smoking-induced vasculopathy and to evaluate the targets of its effects., Main Methods: Nicotine was orally administered for 28 days. Thereafter, the rats were orally treated with melatonin for another 28 days. Vessel damage, an important vasoconstrictor peptide (endothelin-1) and the oxidative stress markers were analysed., Key Findings: Nicotine treatment induced marked endothelial damage and an obvious vasoconstriction in the aorta as evaluated by an increased endothelin-1 (ET-1) expression. These alterations were correlated with a reduction of endothelial nitric oxide synthase (eNOS) and superoxide dismutase (SOD) and with increases of heat shock protein (Hsp70) and inducible nitric oxide synthase (iNOS) activities. Melatonin not only improved the impairment of endothelial-dependent relaxation, but also induced the increase of eNOS and SOD and the reduction of iNOS and Hsp70., Significance: The findings indicate that nicotine is associated with an elevated synthesis of the vasoconstrictor peptide (ET-1); it also induces a reduction of nitric oxide-mediated vasodilatation (eNOS) and promotes oxidative stress in the vessel wall. We propose that melatonin should be considered as a therapeutic intervention for smokers since it reduces vasoconstriction and oxidative stress and improves endothelial physiology., (Copyright © 2010 Elsevier Inc. All rights reserved.)
- Published
- 2010
- Full Text
- View/download PDF
43. Aquaporins and neurodegenerative diseases.
- Author
-
Foglio E and Rodella LF
- Abstract
Aquaporins (AQPs) are a family of widely distributed membrane-inserted water channel proteins providing a pathway for osmotically-driven water, glycerol, urea or ions transport through cell membranes and mechanisms to control particular aspects of homeostasis. Beside their physiological expression patterns in Central Nervous System (CNS), it is conceivable that AQPs are also abnormally expressed in some pathological conditions interesting CNS (e.g. neurodegenerative diseases) in which preservation of brain homeostasis is at risk.The purpose of this review was to take in consideration those neurodegenerative diseases in whose pathogenetic processes it was possible to hypothesize some alterations in CNS AQPs expression or modulation leading to damages of brain water homeostasis.
- Published
- 2010
- Full Text
- View/download PDF
44. Clinical biomarkers in kidney diseases.
- Author
-
Bonomini F, Foglio E, Rodella LF, and Rezzani R
- Subjects
- Acute Kidney Injury classification, Acute Kidney Injury epidemiology, Acute Kidney Injury pathology, Acute-Phase Proteins metabolism, Cystatin C metabolism, Fatty Acid-Binding Proteins metabolism, Hepatitis A Virus Cellular Receptor 1, Humans, Interleukin-18 metabolism, Lipocalin-2, Lipocalins metabolism, Membrane Glycoproteins metabolism, Proto-Oncogene Proteins metabolism, Receptors, Virus metabolism, Renal Insufficiency, Chronic pathology, Acute Kidney Injury diagnosis, Acute Kidney Injury metabolism, Biomarkers metabolism, Renal Insufficiency, Chronic diagnosis, Renal Insufficiency, Chronic metabolism
- Abstract
Biomarkers are "biological parameters that can be objectively measured and evaluated, which act as indicators of normal or pathogenic processes, or of the pharmacological response to a therapeutic intervention". Renal failure can be broadly divided in acute and chronic renal diseases, two classes of renal pathology that are well distinct each other, not only on the basis of duration and reversibility of loss of kidney function, but also because of their different aetiopathological processes and their different histopathological characteristics. Unlikely, the conventional measures used for monitoring kidney function are not ideal in the diagnosis of neither acute or chronic kidney diseases and has impaired our ability to institute potentially effective therapies.Therefore, researchers are seeking new early, predictive, non-invasive biomarkers that can aid in the diagnosis for both acute and chronic diseases.These biomarkers will be useful for assessing the duration and severity of kidney disease, and for predicting progression and adverse clinical outcomes.This review article summarized our current understanding of the acute and chronic renal diseases and discussed the most promising biomarkers for facilitating early detection and predicting clinical outcomes.
- Published
- 2010
- Full Text
- View/download PDF
45. Role of heme oxygenase in modulating endothelial function in mesenteric small resistance arteries of spontaneously hypertensive rats.
- Author
-
Porteri E, Rodella LF, Rezzani R, Rizzoni D, Paiardi S, de Ciuceis C, Boari GE, Foglio E, Favero G, Rizzardi N, Platto C, and Agabiti Rosei E
- Subjects
- Acetylcholine pharmacology, Animals, Bradykinin pharmacology, Endothelium, Vascular drug effects, Enzyme Activation drug effects, Enzyme Inhibitors pharmacology, Heme Oxygenase (Decyclizing) antagonists & inhibitors, Immunohistochemistry, In Vitro Techniques, Mesenteric Arteries drug effects, Metalloporphyrins pharmacology, Nitric Oxide physiology, Oxidative Stress, Protoporphyrins pharmacology, Rats, Rats, Inbred SHR, Rats, Inbred WKY, Vascular Resistance, Vasodilation drug effects, omega-N-Methylarginine pharmacology, Endothelium, Vascular enzymology, Endothelium, Vascular physiopathology, Heme Oxygenase (Decyclizing) physiology, Hypertension enzymology, Hypertension physiopathology, Mesenteric Arteries enzymology, Mesenteric Arteries physiopathology
- Abstract
It has been proposed that endothelial dysfunction is due to the excessive degradation of nitric oxide (NO) by oxidative stress. The enzyme heme-oxygenase (HO) seems to exert a protective effect on oxidative stress in the vasculature, both in animal models and in humans. The objective of this study is to evaluate the effects of inhibition or activation of HO on endothelial function in mesenteric small resistance arteries of spontaneously hypertensive rats (SHR). Six SHR were treated with cobalt protoporphyrin IX 50 mg/Kg (CoPP), an activator of HO; six SHR with stannous mesoporphyrin 30 mg/Kg (SnMP), an inhibitor of HO, and six SHR with saline. As controls, six Wistar-Kyoto rats (WKY) were treated with CoPP, six WKY with SnMP, and six WKY with saline. Drugs were injected in the peritoneum once a week for 2 weeks. Systolic blood pressure (SBP) was measured (tail cuff method) before and after treatment. Mesenteric small resistance arteries were mounted on a micromyograph. Endothelial function was evaluated as a cumulative concentration-response curve to acetylcholine (ACH), before and after preincubation with N(G)-methyl-L-arginine (L-NMMA, inhibitor of NO synthase), and to bradykinin (BK). In SHR treatment with CoPP, improved ACH-and BK-induced vasodilatation (ANOVA p < 0.001) and this improvement was abolished by L-NMMA (ANOVA p < 0.001). SnMP was devoid of effects on endothelial function. In WKY, both activation and inhibition of HO did not substantially affect endothelium-mediated vasodilatation. The stimulation of HO seems to induce an improvement of endothelial dysfunction in SHR by possibly reducing oxidative stress and increasing NO availability.
- Published
- 2009
- Full Text
- View/download PDF
46. Aluminium exposure induces Alzheimer's disease-like histopathological alterations in mouse brain.
- Author
-
Rodella LF, Ricci F, Borsani E, Stacchiotti A, Foglio E, Favero G, Rezzani R, Mariani C, and Bianchi R
- Subjects
- Aluminum toxicity, Alzheimer Disease metabolism, Alzheimer Disease pathology, Amyloid beta-Peptides metabolism, Animals, Brain metabolism, Cerebellar Cortex metabolism, Disease Models, Animal, Endoplasmic Reticulum Chaperone BiP, Heat-Shock Proteins metabolism, Humans, Immunohistochemistry, Male, Mice, Mice, Inbred C57BL, Molecular Chaperones metabolism, Neurons metabolism, Plaque, Amyloid chemistry, Toxicity Tests, Chronic methods, Aluminum administration & dosage, Alzheimer Disease chemically induced, Brain pathology
- Abstract
Aluminium (Al) is a neurotoxic metal and Al exposure may be a factor in the aetiology of various neurodegenerative diseases such as Alzheimer's disease (AD). The major pathohistological findings in the AD brain are the presence of neuritic plaques containing beta-amyloid (Abeta) which may interfere with neuronal communication. Moreover, it has been observed that GRP78, a stress-response protein induced by conditions that adversely affect endoplasmic reticulum (ER) function, is reduced in the brain of AD patients. In this study, we investigated the correlation between the expression of Abeta and GRP78 in the brain cortex of mice chronically treated with aluminium sulphate. Chronic exposure over 12 months to aluminium sulphate in drinking water resulted in deposition of Abeta similar to that seen in congophilic amyloid angiopathy (CAA) in humans and a reduction in neuronal expression of GRP78 similar to what has previously been observed in Alzheimer's disease. So, we hypothesise that chronic Al administration is responsible for oxidative cell damage that interferes with ER functions inducing Abeta accumulation and neurodegenerative damage.
- Published
- 2008
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.