Network


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

Hotspot


Dive into the research topics where Francesca Favaretto is active.

Publication


Featured researches published by Francesca Favaretto.


Human Mutation | 2015

Alström Syndrome: Mutation Spectrum of ALMS1

Jan D. Marshall; Jean Muller; Gayle B. Collin; Gabriella Milan; Stephen F. Kingsmore; Darrell L. Dinwiddie; Emily Farrow; Neil Miller; Francesca Favaretto; Pietro Maffei; Hélène Dollfus; Roberto Vettor; Juergen K Naggert

Alström Syndrome (ALMS), a recessive, monogenic ciliopathy caused by mutations in ALMS1, is typically characterized by multisystem involvement including early cone‐rod retinal dystrophy and blindness, hearing loss, childhood obesity, type 2 diabetes mellitus, cardiomyopathy, fibrosis, and multiple organ failure. The precise function of ALMS1 remains elusive, but roles in endosomal and ciliary transport and cell cycle regulation have been shown. The aim of our study was to further define the spectrum of ALMS1 mutations in patients with clinical features of ALMS. Mutational analysis in a world‐wide cohort of 204 families identified 109 novel mutations, extending the number of known ALMS1 mutations to 239 and highlighting the allelic heterogeneity of this disorder. This study represents the most comprehensive mutation analysis in patients with ALMS, identifying the largest number of novel mutations in a single study worldwide. Here, we also provide an overview of all ALMS1 mutations identified to date.


AIDS | 2006

Microarray analysis during adipogenesis identifies new genes altered by antiretroviral drugs.

Monia Pacenti; Luisa Barzon; Francesca Favaretto; Karina Fincati; Sara Romano; Gabriella Milan; Roberto Vettor; Giorgio Palù

Objective:To elucidate the pathogenesis of HAART-associated lipodystrophy, by investigating the effects of antiretroviral drugs on adipocyte differentiation and gene expression profile. Design and methods:Analysis of gene expression profile by DNA microarrays and quantitative RT–PCR of 3T3-L1 preadipocytes treated with the nucleoside reverse transcriptase inhibitors (NRTI) lamivudine, zidovudine, stavudine, and zalcitabine, and with the protease inhibitors (PI) indinavir, saquinavir, and lopinavir during maturation into adipocytes. Results:Under standard adipogenic differentiation protocols, PI significantly inhibited adipocyte differentiation, as demonstrated by cell viability assay and Oil Red O staining and quantification, whereas NRTI had mild effects on adipogenesis. Gene expression profile analysis showed that treatment with NRTI modulated the expression of transcription factors, such as Aebp1, Pou5f1 and Phf6, which could play a key role in the determination of the adipocyte phenotype. PI also modulated gene expression toward inhibition of adipocyte differentiation, with up-regulation of the Wnt signaling gene Wnt10a and down-regulation of the expression of genes encoding master adipogenic transcription factors (e.g., C/EBPα and PPARγ), oestrogen receptor β, and adipocyte-specific markers (e.g., Adiponectin, Leptin, Mrap, Cd36, S100A8). Conclusions:This study identifies new genes modulated by PI and NRTI in differentiating adipocytes. Abnormal expression of these genes, which include master adipogenic transcription factors and genes involved in lipid metabolism and cell cycle control, could contribute to the understanding of the pathogenesis of HAART-associated lipodystrophy.


PLOS ONE | 2011

ALMS1-deficient fibroblasts over-express extra-cellular matrix components, display cell cycle delay and are resistant to apoptosis.

Elisabetta Zulato; Francesca Favaretto; Caterina Veronese; Stefano Campanaro; Jan D. Marshall; Sara Romano; Anna Cabrelle; Gayle B. Collin; Barbara Zavan; Anna S. Belloni; Enrica Rampazzo; Jürgen K. Naggert; Giovanni Abatangelo; Nicola Sicolo; Pietro Maffei; Gabriella Milan; Roberto Vettor

Alström Syndrome (ALMS) is a rare genetic disorder (483 living cases), characterized by many clinical manifestations, including blindness, obesity, type 2 diabetes and cardiomyopathy. ALMS is caused by mutations in the ALMS1 gene, encoding for a large protein with implicated roles in ciliary function, cellular quiescence and intracellular transport. Patients with ALMS have extensive fibrosis in nearly all tissues resulting in a progressive organ failure which is often the ultimate cause of death. To focus on the role of ALMS1 mutations in the generation and maintenance of this pathological fibrosis, we performed gene expression analysis, ultrastructural characterization and functional assays in 4 dermal fibroblast cultures from ALMS patients. Using a genome-wide gene expression analysis we found alterations in genes belonging to specific categories (cell cycle, extracellular matrix (ECM) and fibrosis, cellular architecture/motility and apoptosis). ALMS fibroblasts display cytoskeleton abnormalities and migration impairment, up-regulate the expression and production of collagens and despite the increase in the cell cycle length are more resistant to apoptosis. Therefore ALMS1-deficient fibroblasts showed a constitutively activated myofibroblast phenotype even if they do not derive from a fibrotic lesion. Our results support a genetic basis for the fibrosis observed in ALMS and show that both an excessive ECM production and a failure to eliminate myofibroblasts are key mechanisms. Furthermore, our findings suggest new roles for ALMS1 in both intra- and extra-cellular events which are essential not only for the normal cellular function but also for cell-cell and ECM-cell interactions.


PLOS ONE | 2014

GLUT4 Defects in Adipose Tissue Are Early Signs of Metabolic Alterations in Alms1GT/GT, a Mouse Model for Obesity and Insulin Resistance

Francesca Favaretto; Gabriella Milan; Gayle B. Collin; Jan D. Marshall; Fabio Stasi; Pietro Maffei; Roberto Vettor; Juergen K Naggert

Dysregulation of signaling pathways in adipose tissue leading to insulin resistance can contribute to the development of obesity-related metabolic disorders. Alström Syndrome, a recessive ciliopathy, caused by mutations in ALMS1, is characterized by progressive metabolic alterations such as childhood obesity, hyperinsulinemia, and type 2 diabetes. Here we investigated the role of Alms1 disruption in AT expansion and insulin responsiveness in a murine model for Alström Syndrome. A gene trap insertion in Alms1 on the insulin sensitive C57BL6/Ei genetic background leads to early hyperinsulinemia and a progressive increase in body weight. At 6 weeks of age, before the onset of the metabolic disease, the mutant mice had enlarged fat depots with hypertrophic adipocytes, but without signs of inflammation. Expression of lipogenic enzymes was increased. Pre-adipocytes isolated from mutant animals demonstrated normal adipogenic differentiation but gave rise to mature adipocytes with reduced insulin-stimulated glucose uptake. Assessment of whole body glucose homeostasis revealed glucose intolerance. Insulin stimulation resulted in proper AKT phosphorylation in adipose tissue. However, the total amount of glucose transporter 4 (SLC4A2) and its translocation to the plasma membrane were reduced in mutant adipose depots compared to wildtype littermates. Alterations in insulin stimulated trafficking of glucose transporter 4 are an early sign of metabolic dysfunction in Alström mutant mice, providing a possible explanation for the reduced glucose uptake and the compensatory hyperinsulinemia. The metabolic signaling deficits either reside downstream or are independent of AKT activation and suggest a role for ALMS1 in GLUT4 trafficking. Alström mutant mice represent an interesting model for the development of metabolic disease in which adipose tissue with a reduced glucose uptake can expand by de novo lipogenesis to an obese state.


Pediatric Diabetes | 2012

The progression from obesity to type 2 diabetes in Alström syndrome.

Vera Bettini; Pietro Maffei; Claudio Pagano; Sara Romano; Gabriella Milan; Francesca Favaretto; Jan D. Marshall; Richard B Paisey; Francesco Scolari; Nella Greggio; Ilaria Tosetto; Jürgen K. Naggert; Nicola Sicolo; Roberto Vettor

Bettini V, Maffei P, Pagano C, Romano S, Milan G, Favaretto F, Marshall JD, Paisey R, Scolari F, Greggio NA, Tosetto I, Naggert JK, Sicolo N, Vettor R. The progression from obesity to type 2 diabetes in Alström syndrome.


Clinical Endocrinology | 2013

Alström syndrome is associated with short stature and reduced GH reserve

Sara Romano; Pietro Maffei; Bettini; Gabriella Milan; Francesca Favaretto; Marina Gardiman; Jd Marshall; Na Greggio; Gb Pozzan; Gb Collin; Jk Naggert; Roderick T. Bronson; Roberto Vettor

Alström syndrome (ALMS) is a rare autosomal recessive monogenic disease included in an emerging class of genetic disorders called ‘ciliopathies’ and is likely to impact the central nervous system as well as metabolic and endocrine function. Individuals with ALMS present clinical features resembling a growth hormone deficiency (GHD) condition, but thus far no study has specifically investigated this aspect in a large population.


Clinical Lipidology | 2009

Adipogenic potential of skeletal muscle satellite cells

Marta Sanna; Chiara Franzin; Michela Pozzobon; Francesca Favaretto; Carlo Alberto Rossi; Alessandra Calcagno; Alessandro Scarda; Chiara Dal Pra; Catia Pilon; Gabriella Milan; Giovanni Federspil; Paolo De Coppi; Roberto Vettor

Abstract Satellite cells (SCs) are undifferentiated skeletal muscle cells that allow muscular growth and regeneration. Since their first description, SCs were simply considered myogenic precursors, but now it is widely accepted that SCs are a heterogeneous stem cell population characterized by plasticity and self-renewal. In this report we focus on SCs capacity to undergo adipogenic differentiation both spontaneous and induced by adipogenic factors. Understanding SC behavior is especially important because their adipogenic potential could represent a pathophysiological explanation for the intramuscular fat depots and associated insulin resistance that characterize many metabolic diseases and age-related sarcopenia. Moreover, SC are a therapeutic promise for neuromuscular diseases in the context of tissue engineering, representing an interesting cell source for implantation.


Scientific Reports | 2017

CK2 modulates adipocyte insulin-signaling and is up-regulated in human obesity

Christian Borgo; Gabriella Milan; Francesca Favaretto; Fabio Stasi; Roberto Fabris; Valentina Salizzato; Luca Cesaro; Anna Belligoli; Marta Sanna; Mirto Foletto; Luca Prevedello; Vincenzo Vindigni; Romeo Bardini; Arianna Donella-Deana; Roberto Vettor

Insulin plays a major role in glucose metabolism and insulin-signaling defects are present in obesity and diabetes. CK2 is a pleiotropic protein kinase implicated in fundamental cellular pathways and abnormally elevated in tumors. Here we report that in human and murine adipocytes CK2-inhibition decreases the insulin-induced glucose-uptake by counteracting Akt-signaling and GLUT4-translocation to the plasma membrane. In mice CK2 acts on insulin-signaling in adipose tissue, liver and skeletal muscle and its acute inhibition impairs glucose tolerance. Notably, CK2 protein-level and activity are greatly up-regulated in white adipose tissue from ob/ob and db/db mice as well as from obese patients, regardless the severity of their insulin-resistance and the presence of pre-diabetes or overt type 2 diabetes. Weight loss obtained by both bariatric surgery or hypocaloric diet reverts CK2 hyper-activation to normal level. Our data suggest a central role of CK2 in insulin-sensitivity, glucose homeostasis and adipose tissue remodeling. CK2 up-regulation is identified as a hallmark of adipose tissue pathological expansion, suggesting a new potential therapeutic target for human obesity.


Human Mutation | 2017

Monogenic diabetes syndromes: Locus‐specific databases for Alström, Wolfram, and Thiamine‐responsive megaloblastic anemia

Dewi Astuti; Ataf Sabir; Piers Fulton; Malgorzata Zatyka; Denise Williams; Carol Hardy; Gabriella Milan; Francesca Favaretto; Patrick Yu-Wai-Man; Julia Rohayem; Miguel López de Heredia; Tamara Hershey; Lisbeth Tranebjærg; Jian-Hua Chen; Annabel Chaussenot; Virginia Nunes; Bess A. Marshall; Susan McAfferty; Vallo Tillmann; Pietro Maffei; Véronique Paquis-Flucklinger; Tarekign Geberhiwot; Wojciech Mlynarski; Kay Parkinson; Virginie Picard; Gema Esteban Bueno; Renuka Dias; Amy Arnold; Caitlin Richens; Richard B Paisey

We developed a variant database for diabetes syndrome genes, using the Leiden Open Variation Database platform, containing observed phenotypes matched to the genetic variations. We populated it with 628 published disease‐associated variants (December 2016) for: WFS1 (n = 309), CISD2 (n = 3), ALMS1 (n = 268), and SLC19A2 (n = 48) for Wolfram type 1, Wolfram type 2, Alström, and Thiamine‐responsive megaloblastic anemia syndromes, respectively; and included 23 previously unpublished novel germline variants in WFS1 and 17 variants in ALMS1. We then investigated genotype–phenotype relations for the WFS1 gene. The presence of biallelic loss‐of‐function variants predicted Wolfram syndrome defined by insulin‐dependent diabetes and optic atrophy, with a sensitivity of 79% (95% CI 75%–83%) and specificity of 92% (83%–97%). The presence of minor loss‐of‐function variants in WFS1 predicted isolated diabetes, isolated deafness, or isolated congenital cataracts without development of the full syndrome (sensitivity 100% [93%–100%]; specificity 78% [73%–82%]). The ability to provide a prognostic prediction based on genotype will lead to improvements in patient care and counseling. The development of the database as a repository for monogenic diabetes gene variants will allow prognostic predictions for other diabetes syndromes as next‐generation sequencing expands the repertoire of genotypes and phenotypes. The database is publicly available online at https://lovd.euro-wabb.org.


American Journal of Physiology-endocrinology and Metabolism | 2017

Increased mitochondrial calcium uniporter in adipocytes underlies mitochondrial alterations associated with insulin resistance

Lauren E. Wright; Denis Vecellio Reane; Gabriella Milan; Anna Terrin; Giorgia Di Bello; Anna Belligoli; Marta Sanna; Mirto Foletto; Francesca Favaretto; Anna Raffaello; Cristina Mammucari; Donato Nitti; Roberto Vettor; Rosario Rizzuto

Intracellular calcium influences an array of pathways and affects cellular processes. With the rapidly progressing research investigating the molecular identity and the physiological roles of the mitochondrial calcium uniporter (MCU) complex, we now have the tools to understand the functions of mitochondrial Ca2+ in the regulation of pathophysiological processes. Herein, we describe the role of key MCU complex components in insulin resistance in mouse and human adipose tissue. Adipose tissue gene expression was analyzed from several models of obese and diabetic rodents and in 72 patients with obesity as well as in vitro insulin-resistant adipocytes. Genetic manipulation of MCU activity in 3T3-L1 adipocytes allowed the investigation of the role of mitochondrial calcium uptake. In insulin-resistant adipocytes, mitochondrial calcium uptake increased and several MCU components were upregulated. Similar results were observed in mouse and human visceral adipose tissue (VAT) during the progression of obesity and diabetes. Intriguingly, subcutaneous adipose tissue (SAT) was spared from overt MCU fluctuations. Furthermore, MCU expression returned to physiological levels in VAT of patients after weight loss by bariatric surgery. Genetic manipulation of mitochondrial calcium uptake in 3T3-L1 adipocytes demonstrated that changes in mitochondrial calcium concentration ([Ca2+]mt) can affect mitochondrial metabolism, including oxidative enzyme activity, mitochondrial respiration, membrane potential, and reactive oxygen species formation. Finally, our data suggest a strong relationship between [Ca2+]mt and the release of IL-6 and TNFα in adipocytes. Altered mitochondrial calcium flux in fat cells may play a role in obesity and diabetes and may be associated with the differential metabolic profiles of VAT and SAT.

Collaboration


Dive into the Francesca Favaretto's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge