Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Maria Mavilio is active.

Publication


Featured researches published by Maria Mavilio.


Embo Molecular Medicine | 2013

Loss of TIMP3 underlies diabetic nephropathy via FoxO1/STAT1 interplay.

Loredana Fiorentino; Michele Cavalera; Stefano Menini; Valentina Marchetti; Maria Mavilio; Marta Fabrizi; Francesca Conserva; Viviana Casagrande; Rossella Menghini; Paola Pontrelli; Ivan Arisi; Mara D'Onofrio; Davide Lauro; Rama Khokha; Domenico Accili; Giuseppe Pugliese; Loreto Gesualdo; Renato Lauro; Massimo Federici

ADAM17 and its inhibitor TIMP3 are involved in nephropathy, but their role in diabetic kidney disease (DKD) is unclear. Diabetic Timp3−/− mice showed increased albuminuria, increased membrane thickness and mesangial expansion. Microarray profiling uncovered a significant reduction of Foxo1 expression in diabetic Timp3−/− mice compared to WT, along with FoxO1 target genes involved in autophagy, while STAT1, a repressor of FoxO1 transcription, was increased. Re‐expression of Timp3 in Timp3−/− mesangial cells rescued the expression of Foxo1 and its targets, and decreased STAT1 expression to control levels; abolishing STAT1 expression led to a rescue of FoxO1, evoking a role of STAT1 in linking Timp3 deficiency to FoxO1. Studies on kidney biopsies from patients with diabetic nephropathy confirmed a significant reduction in TIMP3, FoxO1 and FoxO1 target genes involved in autophagy compared to controls, while STAT1 expression was strongly increased.


Acta Diabetologica | 2013

Regulation of TIMP3 in diabetic nephropathy: a role for microRNAs

Loredana Fiorentino; Michele Cavalera; Maria Mavilio; Francesca Conserva; Rossella Menghini; Loreto Gesualdo; Massimo Federici

Diabetic nephropathy (DN) is the major cause of chronic kidney disease in developed countries and contributes significantly to increased morbidity and mortality among diabetic patients. Morphologically, DN is characterized by tubulo-interstitial fibrosis, thickening of the glomerular basement membrane and mesangial expansion mainly due to accumulation of extracellular matrix (ECM). ECM turnover is regulated by metalloproteinases and tissue inhibitors of metalloproteinases (TIMPs) activities. In diabetic conditions, TIMP3 expression in kidney is strongly reduced, but the causes of this reduction are still unknown. The aim of this study was to elucidate at least one of these mechanisms which relies on differential expression of TIMP3-targeting microRNAs (miRs) in a hyperglycemic environment either in vitro (MES13 cell line) or in vivo (mouse kidney and human biopsies). Among the TIMP3-targeting miRs, miR-21 and miR-221 were significantly upregulated in kidneys from diabetic mice compared to control littermates, and in a mesangial cell line grown in high glucose conditions. In human samples, only miR-21 expression was increased in kidney biopsies from diabetic patients compared to healthy controls. The expression of miR-217, which targets TIMP3 indirectly through downregulation of SirT1, was also increased in diabetic kidney and MES13 cell line. In agreement with these result, SirT1 expression was reduced in mouse and human diabetic kidneys as well as in MES13 mesangial cell line. TIMP3 deficiency has recently emerged as a hallmark of DN in mouse and human. In this study, we demonstrated that this reduction is due, at least in part, to increased expression of certain TIMP3-targeting miRs in diabetic kidneys compared to healthy controls. Unveiling the post-transcriptional mechanisms responsible for TIMP3 downregulation in hyperglycemic conditions may orient toward the use of this protein as a possible therapeutic target in DN.


Diabetes | 2014

IL-21 is a major negative regulator of IRF4-dependent lipolysis affecting Tregs in adipose tissue and systemic insulin sensitivity.

Marta Fabrizi; Valentina Marchetti; Maria Mavilio; Arianna Marino; Viviana Casagrande; Michele Cavalera; José Maria Moreno Navarrete; Teresa Mezza; Gian Pio Sorice; Loredana Fiorentino; Rossella Menghini; Renato Lauro; Giovanni Monteleone; Andrea Giaccari; José Manuel Fernández Real; Massimo Federici

Obesity elicits immune cell infiltration of adipose tissue provoking chronic low-grade inflammation. Regulatory T cells (Tregs) are specifically reduced in adipose tissue of obese animals. Since interleukin (IL)-21 plays an important role in inducing and maintaining immune-mediated chronic inflammatory processes and negatively regulates Treg differentiation/activity, we hypothesized that it could play a role in obesity-induced insulin resistance. We found IL-21 and IL-21R mRNA expression upregulated in adipose tissue of high-fat diet (HFD) wild-type (WT) mice and in stromal vascular fraction from human obese subjects in parallel to macrophage and inflammatory markers. Interestingly, a larger infiltration of Treg cells was seen in the adipose tissue of IL-21 knockout (KO) mice compared with WT animals fed both normal diet and HFD. In a context of diet-induced obesity, IL-21 KO mice, compared with WT animals, exhibited lower body weight, improved insulin sensitivity, and decreased adipose and hepatic inflammation. This metabolic phenotype is accompanied by a higher induction of interferon regulatory factor 4 (IRF4), a transcriptional regulator of fasting lipolysis in adipose tissue. Our data suggest that IL-21 exerts negative regulation on IRF4 and Treg activity, developing and maintaining adipose tissue inflammation in the obesity state.


Atherosclerosis | 2014

Loss of TIMP3 exacerbates atherosclerosis in ApoE null mice

Robert Stöhr; Michele Cavalera; Stefano Menini; Maria Mavilio; Viviana Casagrande; Claudia Rossi; Andrea Urbani; Marina Cardellini; Giuseppe Pugliese; Rossella Menghini; Massimo Federici

BACKGROUND Tissue inhibitor of metalloproteinase 3 (TIMP3) is a stromal protein that inhibits the activity of various proteases and receptors. We have previously shown TIMP3 to be downregulated in metabolic and inflammatory disorders, such as type 2 diabetes mellitus. We have now generated an ApoE(-/-)Timp3(-/-) mouse model in which, through the use of genetics, metabolomics and in-vivo phenotypical analysis we investigated the role of TIMP3 in the development of atherosclerosis. METHODS AND RESULTS En face aorta analysis and aortic root examination showed that ApoE(-/-)Timp3(-/-) mice show increased atherosclerosis with increased infiltration of macrophages into the plaque. Serum concentration of MCP-1 were elevated in the serum of ApoE(-/-)Timp3(-/-) mice coupled with an expansion of the inflammatory (M1) Gr1+ macrophages, both in the circulation and within the aortic tissue. Targeted analysis of metabolites revealed a trend to reduced short chain acylcarnitines. CONCLUSIONS Our study shows that lack of TIMP3 increases inflammation and polarizes macrophages towards a more inflammatory phenotype resulting in increased atherosclerosis.


Diabetes | 2014

ITCH Deficiency Protects From Diet-Induced Obesity

Arianna Marino; Rossella Menghini; Marta Fabrizi; Viviana Casagrande; Maria Mavilio; Robert Stoehr; Eleonora Candi; Alessandro Mauriello; José María Moreno-Navarrete; María Gómez-Serrano; Belén Peral; Gerry Melino; Renato Lauro; José Manuel Fernández Real; Massimo Federici

Classically activated macrophages (M1) secrete proinflammatory cytokine and are predominant in obese adipose tissue. M2 macrophages, prevalent in lean adipose tissue, are induced by IL-13 and IL-4, mainly secreted by Th2 lymphocytes, and produce the anti-inflammatory cytokine IL-10. ITCH is a ubiquitously expressed E3 ubiquitin ligase involved in T-cell differentiation and in a wide range of inflammatory pathways. ITCH downregulation in lymphocytes causes aberrant Th2 differentiation. To investigate the role of Th2/M2 polarization in obesity-related inflammation and insulin resistance, we compared wild-type and Itch−/− mice in a context of diet-induced obesity (high-fat diet [HFD]). When subjected to HFD, Itch−/− mice did not show an increase in body weight or insulin resistance; calorimetric analysis suggested an accelerated metabolism. The molecular analysis of metabolically active tissue revealed increased levels of M2 markers and genes involved in fatty acid oxidation. Histological examination of livers from Itch−/− mice suggested that ITCH deficiency protects mice from obesity-related nonalcoholic fatty liver disease. We also found a negative correlation between ITCH and M2 marker expression in human adipose tissues. Taken together, our data indicate that ITCH E3 ubiquitin ligase deficiency protects from the metabolic disorder caused by obesity.


Scientific Reports | 2015

ITCH modulates SIRT6 and SREBP2 to influence lipid metabolism and atherosclerosis in ApoE null mice.

Robert Stöhr; Maria Mavilio; Arianna Marino; Casagrande; Ben Arpad Kappel; Julia Möllmann; Rossella Menghini; Gerry Melino; Massimo Federici

Atherosclerosis is a chronic inflammatory disease characterized by the infiltration of pro-inflammatory macrophages into a lipid-laden plaque. ITCH is an E3 ubiquitin ligase that has been shown to polarize macrophages to an anti-inflammatory phenotype. We therefore investigated the effect of ITCH deficiency on the development of atherosclerosis. ApoE−/−ITCH−/− mice fed a western diet for 12 weeks showed increased circulating M2 macrophages together with a reduction in plaque formation. Bone marrow transplantation recreated the haemopoietic phenotype of increased circulating M2 macrophages but failed to affect plaque development. Intriguingly, the loss of ITCH lead to a reduction in circulating cholesterol levels through interference with nuclear SREBP2 clearance. This resulted in increased LDL reuptake through upregulation of LDL receptor expression. Furthermore, ApoE−/−ITCH−/− mice exhibit reduced hepatic steatosis, increased mitochondrial oxidative capacity and an increased reliance on fatty acids as energy source. We found that ITCH ubiquitinates SIRT6, leading to its breakdown, and thus promoting hepatic lipid infiltration through reduced fatty acid oxidation. The E3 Ubiquitin Ligase ITCH modulates lipid metabolism impacting on atherosclerosis progression independently from effects on myeloid cells polarization through control of SIRT6 and SREBP2 ubiquitination. Thus, modulation of ITCH may provide a target for the treatment of hypercholesterolemia and hyperlipidemia.


Molecular metabolism | 2015

TIMP3 interplays with apelin to regulate cardiovascular metabolism in hypercholesterolemic mice.

Robert Stöhr; Ben Arpad Kappel; Daniela Carnevale; Michele Cavalera; Maria Mavilio; Ivan Arisi; Valentina Fardella; Giuseppe Cifelli; Viviana Casagrande; Stefano Rizza; Antonino Cattaneo; Alessandro Mauriello; Rossella Menghini; Giuseppe Lembo; Massimo Federici

Objective Tissue inhibitor of metalloproteinase 3 (TIMP3) is an extracellular matrix (ECM) bound protein, which has been shown to be downregulated in human subjects and experimental models with cardiometabolic disorders, including type 2 diabetes mellitus, hypertension and atherosclerosis. The aim of this study was to investigate the effects of TIMP3 on cardiac energy homeostasis during increased metabolic stress conditions. Methods ApoE−/−TIMP3−/− and ApoE−/− mice on a C57BL/6 background were subjected to telemetric ECG analysis and experimental myocardial infarction as models of cardiac stress induction. We used Western blot, qRT-PCR, histology, metabolomics, RNA-sequencing and in vivo phenotypical analysis to investigate the molecular mechanisms of altered cardiac energy metabolism. Results ApoE−/−TIMP3−/− revealed decreased lifespan. Telemetric ECG analysis showed increased arrhythmic episodes, and experimental myocardial infarction by left anterior descending artery (LAD) ligation resulted in increased peri-operative mortality together with increased scar formation, ventricular dilatation and a reduction of cardiac function after 4 weeks in the few survivors. Hearts of ApoE−/−TIMP3−/− exhibited accumulation of neutral lipids when fed a chow diet, which was exacerbated by a high fat, high cholesterol diet. Metabolomics analysis revealed an increase in circulating markers of oxidative stress with a reduction in long chain fatty acids. Using whole heart mRNA sequencing, we identified apelin as a putative modulator of these metabolic defects. Apelin is a regulator of fatty acid oxidation, and we found a reduction in the levels of enzymes involved in fatty acid oxidation in the left ventricle of ApoE−/−TIMP3−/− mice. Injection of apelin restored the hitherto identified metabolic defects of lipid oxidation. Conclusion TIMP3 regulates lipid metabolism as well as oxidative stress response via apelin. These findings therefore suggest that TIMP3 maintains metabolic flexibility in the heart, particularly during episodes of increased cardiac stress.


Cell Reports | 2016

A Role for Timp3 in Microbiota-Driven Hepatic Steatosis and Metabolic Dysfunction

Maria Mavilio; Valentina Marchetti; Marta Fabrizi; Robert Stöhr; Arianna Marino; Viviana Casagrande; Loredana Fiorentino; Marina Cardellini; Ben Arpad Kappel; Ivan Monteleone; Celine Garret; Alessandro Mauriello; Giovanni Monteleone; Alessio Farcomeni; Remy Burcelin; Rossella Menghini; Massimo Federici

The effect of gut microbiota on obesity and insulin resistance is now recognized, but the underlying host-dependent mechanisms remain poorly undefined. We find that tissue inhibitor of metalloproteinase 3 knockout (Timp3(-/-)) mice fed a high-fat diet exhibit gut microbiota dysbiosis, an increase in branched chain and aromatic (BCAA) metabolites, liver steatosis, and an increase in circulating soluble IL-6 receptors (sIL6Rs). sIL6Rs can then activate inflammatory cells, such as CD11c(+) cells, which drive metabolic inflammation. Depleting the microbiota through antibiotic treatment significantly improves glucose tolerance, hepatic steatosis, and systemic inflammation, and neutralizing sIL6R signaling reduces inflammation, but only mildly impacts glucose tolerance. Collectively, our results suggest that gut microbiota is the primary driver of the observed metabolic dysfunction, which is mediated, in part, through IL-6 signaling. Our findings also identify an important role for Timp3 in mediating the effect of the microbiota in metabolic diseases.


Cell Reports | 2016

Erratum: A Role for Timp3 in Microbiota-Driven Hepatic Steatosis and Metabolic Dysfunction (Cell Reports (2016) 16(3) (731–743) (S2211124716307677) (10.1016/j.celrep.2016.06.027))

Maria Mavilio; Marchetti; Marta Fabrizi; R Stöhr; Arianna Marino; Casagrande; Loredana Fiorentino; Marina Cardellini; Ben Arpad Kappel; Ivan Monteleone; C Garret; Alessandro Mauriello; Giovanni Monteleone; Alessio Farcomeni; R Burcelin; Rossella Menghini; Massimo Federici

Department of Systems Medicine, University of Rome Tor Vergata, 00133 Rome, Italy 5 Research Unit for Multi-Factorial Diseases, Obesity and Diabetes Scientific Directorate, Bambino 6 Gesù Children Hospital, 00146 Rome, Italy 7 3 Department of Internal Medicine I, University Hospital Aachen, D-52074 Aachen, Germany 8 INSERM U1048, Université Paul Sabatier, IMC, F-31432 Toulouse, France 9 5 Department of Public Health and Infectious Diseases, Sapienza University of Rome, 00161 Rome, 10 Italy 11 *Drs. Mavilio and Marchetti equally contributed 12


Acta Diabetologica | 2018

Proteomic and metabolomic characterization of streptozotocin-induced diabetic nephropathy in TIMP3-deficient mice

Claudia Rossi; Valeria Marzano; Ada Consalvo; Mirco Zucchelli; Stefano Levi Mortera; Viviana Casagrande; Maria Mavilio; Paolo Sacchetta; Massimo Federici; Rossella Menghini; Andrea Urbani; Domenico Ciavardelli

AbstractAimsThe tissue inhibitor of metalloproteinase TIMP3 is a stromal protein that restrains the activity of both protease and receptor in the extracellular matrix and has been found to be down-regulated in diabetic nephropathy (DN), the leading cause of end-stage renal disease in developed countries. MethodsIn order to gain deeper insights on the association of loss of TIMP3 and DN, we performed differential proteomic analysis of kidney and blood metabolic profiling of wild-type and Timp3-knockout mice before and after streptozotocin (STZ) treatment, widely used to induce insulin deficiency and hyperglycemia. ResultsKidney proteomic data and blood metabolic profiles suggest significant alterations of peroxisomal and mitochondrial fatty acids β-oxidation in Timp3-knockout mice compared to wild-type mice under basal condition. These alterations were exacerbated in response to STZ treatment.ConclusionsProteomic and metabolomic approaches showed that loss of TIMP3 alone or in combination with STZ treatment results in significant alterations of kidney lipid metabolism and peripheral acylcarnitine levels, supporting the idea that loss of TIMP3 may generate a phenotype more prone to DN.

Collaboration


Dive into the Maria Mavilio's collaboration.

Top Co-Authors

Avatar

Massimo Federici

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Rossella Menghini

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Viviana Casagrande

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Michele Cavalera

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Ben Arpad Kappel

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Robert Stöhr

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Alessandro Mauriello

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Arianna Marino

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Loredana Fiorentino

University of Rome Tor Vergata

View shared research outputs
Top Co-Authors

Avatar

Marina Cardellini

University of Rome Tor Vergata

View shared research outputs
Researchain Logo
Decentralizing Knowledge