Network


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

Hotspot


Dive into the research topics where Silvia De Rubeis is active.

Publication


Featured researches published by Silvia De Rubeis.


Cell | 2008

The Fragile X Syndrome Protein Represses Activity-Dependent Translation through CYFIP1, a New 4E-BP

Ilaria Napoli; Valentina Mercaldo; Pietro Pilo Boyl; Boris Eleuteri; Francesca Zalfa; Silvia De Rubeis; Daniele Di Marino; Evita Mohr; Marzia Massimi; Mattia Falconi; Walter Witke; Mauro Costa-Mattioli; Nahum Sonenberg; Tilmann Achsel; Claudia Bagni

Strong evidence indicates that regulated mRNA translation in neuronal dendrites underlies synaptic plasticity and brain development. The fragile X mental retardation protein (FMRP) is involved in this process; here, we show that it acts by inhibiting translation initiation. A binding partner of FMRP, CYFIP1/Sra1, directly binds the translation initiation factor eIF4E through a domain that is structurally related to those present in 4E-BP translational inhibitors. Brain cytoplasmic RNA 1 (BC1), another FMRP binding partner, increases the affinity of FMRP for the CYFIP1-eIF4E complex in the brain. Levels of proteins encoded by known FMRP target mRNAs are increased upon reduction of CYFIP1 in neurons. Translational repression is regulated in an activity-dependent manner because BDNF or DHPG stimulation of neurons causes CYFIP1 to dissociate from eIF4E at synapses, thereby resulting in protein synthesis. Thus, the translational repression activity of FMRP in the brain is mediated, at least in part, by CYFIP1.


Nature Neuroscience | 2007

A new function for the fragile X mental retardation protein in regulation of PSD-95 mRNA stability

Francesca Zalfa; Boris Eleuteri; Kirsten S. Dickson; Valentina Mercaldo; Silvia De Rubeis; Alessandra di Penta; Elisabetta Tabolacci; Pietro Chiurazzi; Giovanni Neri; Seth G. N. Grant; Claudia Bagni

Fragile X syndrome (FXS) results from the loss of the fragile X mental retardation protein (FMRP), an RNA-binding protein that regulates a variety of cytoplasmic mRNAs. FMRP regulates mRNA translation and may be important in mRNA localization to dendrites. We report a third cytoplasmic regulatory function for FMRP: control of mRNA stability. In mice, we found that FMRP binds, in vivo, the mRNA encoding PSD-95, a key molecule that regulates neuronal synaptic signaling and learning. This interaction occurs through the 3′ untranslated region of the PSD-95 (also known as Dlg4) mRNA, increasing message stability. Moreover, stabilization is further increased by mGluR activation. Although we also found that the PSD-95 mRNA is synaptically localized in vivo, localization occurs independently of FMRP. Through our functional analysis of this FMRP target we provide evidence that dysregulation of mRNA stability may contribute to the cognitive impairments in individuals with FXS.


Neuron | 2013

CYFIP1 Coordinates mRNA Translation and Cytoskeleton Remodeling to Ensure Proper Dendritic Spine Formation

Silvia De Rubeis; Emanuela Pasciuto; Ka Wan Li; Esperanza Fernández; Daniele Di Marino; Andrea Buzzi; Linnaea E. Ostroff; Eric Klann; Fried J. T. Zwartkruis; Noboru H. Komiyama; Seth G. N. Grant; Christel Poujol; Daniel Choquet; Tilmann Achsel; Danielle Posthuma; August B. Smit; Claudia Bagni

Summary The CYFIP1/SRA1 gene is located in a chromosomal region linked to various neurological disorders, including intellectual disability, autism, and schizophrenia. CYFIP1 plays a dual role in two apparently unrelated processes, inhibiting local protein synthesis and favoring actin remodeling. Here, we show that brain-derived neurotrophic factor (BDNF)-driven synaptic signaling releases CYFIP1 from the translational inhibitory complex, triggering translation of target mRNAs and shifting CYFIP1 into the WAVE regulatory complex. Active Rac1 alters the CYFIP1 conformation, as demonstrated by intramolecular FRET, and is key in changing the equilibrium of the two complexes. CYFIP1 thus orchestrates the two molecular cascades, protein translation and actin polymerization, each of which is necessary for correct spine morphology in neurons. The CYFIP1 interactome reveals many interactors associated with brain disorders, opening new perspectives to define regulatory pathways shared by neurological disabilities characterized by spine dysmorphogenesis.


PLOS Genetics | 2013

Integrated Model of De Novo and Inherited Genetic Variants Yields Greater Power to Identify Risk Genes

Xin He; Stephan J. Sanders; Li Liu; Silvia De Rubeis; Elaine T. Lim; James S. Sutcliffe; Gerard D. Schellenberg; Richard A. Gibbs; Mark J. Daly; Joseph D. Buxbaum; Matthew W. State; Bernie Devlin; Kathryn Roeder

De novo mutations affect risk for many diseases and disorders, especially those with early-onset. An example is autism spectrum disorders (ASD). Four recent whole-exome sequencing (WES) studies of ASD families revealed a handful of novel risk genes, based on independent de novo loss-of-function (LoF) mutations falling in the same gene, and found that de novo LoF mutations occurred at a twofold higher rate than expected by chance. However successful these studies were, they used only a small fraction of the data, excluding other types of de novo mutations and inherited rare variants. Moreover, such analyses cannot readily incorporate data from case-control studies. An important research challenge in gene discovery, therefore, is to develop statistical methods that accommodate a broader class of rare variation. We develop methods that can incorporate WES data regarding de novo mutations, inherited variants present, and variants identified within cases and controls. TADA, for Transmission And De novo Association, integrates these data by a gene-based likelihood model involving parameters for allele frequencies and gene-specific penetrances. Inference is based on a Hierarchical Bayes strategy that borrows information across all genes to infer parameters that would be difficult to estimate for individual genes. In addition to theoretical development we validated TADA using realistic simulations mimicking rare, large-effect mutations affecting risk for ASD and show it has dramatically better power than other common methods of analysis. Thus TADAs integration of various kinds of WES data can be a highly effective means of identifying novel risk genes. Indeed, application of TADA to WES data from subjects with ASD and their families, as well as from a study of ASD subjects and controls, revealed several novel and promising ASD candidate genes with strong statistical support.


Molecular and Cellular Neuroscience | 2010

Fragile X mental retardation protein control of neuronal mRNA metabolism: Insights into mRNA stability.

Silvia De Rubeis; Claudia Bagni

The fragile X mental retardation protein (FMRP) is an RNA binding protein that has an essential role in neurons. From the soma to the synapse, FMRP is associated with a specific subset of messenger RNAs and controls their posttranscriptional fates, i.e., dendritic localization and local translation. Because FMRP target mRNAs encode important neuronal proteins, the deregulation of their expression in the absence of FMRP leads to a strong impairment of synaptic function. Here, we review emerging evidence indicating a critical role for FMRP in the control of mRNA stability. To date, two mRNAs have been identified as being regulated in this manner: PSD-95 mRNA, encoding a scaffolding protein, and Nxf1 mRNA, encoding a general export factor. Moreover, expression studies suggest that the turnover of other neuronal mRNAs, including those encoding for the GABA(A) receptors subunits, could be affected by the loss of FMRP. According to the specific target and/or cellular context, FMRP could influence mRNA stability in the brain.


Human Molecular Genetics | 2015

Genetics and genomics of autism spectrum disorder: embracing complexity

Silvia De Rubeis; Joseph D. Buxbaum

Autism spectrum disorder (ASD) is a neurodevelopmental disorder (NDD) characterized by impairments in social communication and social interaction and the presence of repetitive behaviors and/or restricted interests. ASD has profound etiological and clinical heterogeneity, which has impeded the identification of risk factors and pathophysiological processes underlying the disorder. A constellation of (i) types of genetic variation, (ii) modes of inheritance and (iii) specific genomic loci and genes have all recently been implicated in ASD risk, and these findings are currently being extended with functional analyses in model organisms and genotype-phenotype correlation studies. The overlap of risk loci between ASD and other NDDs raises intriguing questions around the mechanisms of risk. In this review, we will touch upon these aspects of ASD and how they might be addressed.


Journal of Neurodevelopmental Disorders | 2011

Regulation of molecular pathways in the Fragile X Syndrome: insights into Autism Spectrum Disorders.

Silvia De Rubeis; Claudia Bagni

The Fragile X syndrome (FXS) is a leading cause of intellectual disability (ID) and autism. The disease is caused by mutations or loss of the Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein playing multiple functions in RNA metabolism. The expression of a large set of neuronal mRNAs is altered when FMRP is lost, thus causing defects in neuronal morphology and physiology. FMRP regulates mRNA stability, dendritic targeting, and protein synthesis. At synapses, FMRP represses protein synthesis by forming a complex with the Cytoplasmic FMRP Interacting Protein 1 (CYFIP1) and the cap-binding protein eIF4E. Here, we review the clinical, genetic, and molecular aspects of FXS with a special focus on the receptor signaling that regulates FMRP-dependent protein synthesis. We further discuss the FMRP–CYFIP1 complex and its potential relevance for ID and autism.


Advances in Experimental Medicine and Biology | 2012

Molecular and Cellular Aspects of Mental Retardation in The Fragile X Syndrome: from gene mutation/s to spine dysmorphogenesis

Silvia De Rubeis; Esperanza Fernández; Andrea Buzzi; Daniele Di Marino; Claudia Bagni

The Fragile X syndrome (FXS) is the most frequent form of inherited mental retardation and also considered a monogenic cause of Autism Spectrum Disorder. FXS symptoms include neurodevelopmental delay, anxiety, hyperactivity, and autistic-like behavior. The disease is due to mutations or loss of the Fragile X Mental Retardation Protein (FMRP), an RNA-binding protein abundant in the brain and gonads, the two organs mainly affected in FXS patients. FMRP has multiple functions in RNA metabolism, including mRNA decay, dendritic targeting of mRNAs, and protein synthesis. In neurons lacking FMRP, a wide array of mRNAs encoding proteins involved in synaptic structure and function are altered. As a result of this complex dysregulation, in the absence of FMRP, spine morphology and functioning is impaired. Consistently, model organisms for the study of the syndrome recapitulate the phenotype observed in FXS patients, such as dendritic spine anomalies and defects in learning. Here, we review the fundamentals of genetic and clinical aspects of FXS, devoting a specific attention to ASD comorbidity and FXS-related diseases. We also review the current knowledge on FMRP functions through structural, molecular, and cellular findings. Finally, we discuss the neuroanatomical, electrophysiological, and behavioral defects caused by FMRP loss, as well as the current treatments able to partially revert some of the FXS abnormalities.


Nature Neuroscience | 2017

Rates, distribution and implications of postzygotic mosaic mutations in autism spectrum disorder

Elaine T. Lim; Mohammed Uddin; Silvia De Rubeis; Yingleong Chan; Anne S Kamumbu; Xiaochang Zhang; Alissa M. D'Gama; Sonia N Kim; Robert Sean Hill; Arthur P. Goldberg; Christopher S. Poultney; Nancy J. Minshew; Itaru Kushima; Branko Aleksic; Norio Ozaki; Mara Parellada; Celso Arango; Maria Jose Penzol; Angel Carracedo; Alexander Kolevzon; Christina M. Hultman; Lauren A. Weiss; Menachem Fromer; Andreas G. Chiocchetti; Christine M. Freitag; George M. Church; Stephen W. Scherer; Joseph D. Buxbaum; Christopher A. Walsh

We systematically analyzed postzygotic mutations (PZMs) in whole-exome sequences from the largest collection of trios (5,947) with autism spectrum disorder (ASD) available, including 282 unpublished trios, and performed resequencing using multiple independent technologies. We identified 7.5% of de novo mutations as PZMs, 83.3% of which were not described in previous studies. Damaging, nonsynonymous PZMs within critical exons of prenatally expressed genes were more common in ASD probands than controls (P < 1 × 10−6), and genes carrying these PZMs were enriched for expression in the amygdala (P = 5.4 × 10−3). Two genes (KLF16 and MSANTD2) were significantly enriched for PZMs genome-wide, and other PZMs involved genes (SCN2A, HNRNPU and SMARCA4) whose mutation is known to cause ASD or other neurodevelopmental disorders. PZMs constitute a significant proportion of de novo mutations and contribute importantly to ASD risk.


Acta Neuropathologica | 2017

Autism spectrum disorder: neuropathology and animal models

Merina Varghese; Neha Keshav; Sarah Jacot-Descombes; Tahia Warda; Bridget Wicinski; Dara L. Dickstein; Hala Harony-Nicolas; Silvia De Rubeis; Elodie Drapeau; Joseph D. Buxbaum; Patrick R. Hof

Autism spectrum disorder (ASD) has a major impact on the development and social integration of affected individuals and is the most heritable of psychiatric disorders. An increase in the incidence of ASD cases has prompted a surge in research efforts on the underlying neuropathologic processes. We present an overview of current findings in neuropathology studies of ASD using two investigational approaches, postmortem human brains and ASD animal models, and discuss the overlap, limitations, and significance of each. Postmortem examination of ASD brains has revealed global changes including disorganized gray and white matter, increased number of neurons, decreased volume of neuronal soma, and increased neuropil, the last reflecting changes in densities of dendritic spines, cerebral vasculature and glia. Both cortical and non-cortical areas show region-specific abnormalities in neuronal morphology and cytoarchitectural organization, with consistent findings reported from the prefrontal cortex, fusiform gyrus, frontoinsular cortex, cingulate cortex, hippocampus, amygdala, cerebellum and brainstem. The paucity of postmortem human studies linking neuropathology to the underlying etiology has been partly addressed using animal models to explore the impact of genetic and non-genetic factors clinically relevant for the ASD phenotype. Genetically modified models include those based on well-studied monogenic ASD genes (NLGN3, NLGN4, NRXN1, CNTNAP2, SHANK3, MECP2, FMR1, TSC1/2), emerging risk genes (CHD8, SCN2A, SYNGAP1, ARID1B, GRIN2B, DSCAM, TBR1), and copy number variants (15q11-q13 deletion, 15q13.3 microdeletion, 15q11-13 duplication, 16p11.2 deletion and duplication, 22q11.2 deletion). Models of idiopathic ASD include inbred rodent strains that mimic ASD behaviors as well as models developed by environmental interventions such as prenatal exposure to sodium valproate, maternal autoantibodies, and maternal immune activation. In addition to replicating some of the neuropathologic features seen in postmortem studies, a common finding in several animal models of ASD is altered density of dendritic spines, with the direction of the change depending on the specific genetic modification, age and brain region. Overall, postmortem neuropathologic studies with larger sample sizes representative of the various ASD risk genes and diverse clinical phenotypes are warranted to clarify putative etiopathogenic pathways further and to promote the emergence of clinically relevant diagnostic and therapeutic tools. In addition, as genetic alterations may render certain individuals more vulnerable to developing the pathological changes at the synapse underlying the behavioral manifestations of ASD, neuropathologic investigation using genetically modified animal models will help to improve our understanding of the disease mechanisms and enhance the development of targeted treatments.

Collaboration


Dive into the Silvia De Rubeis's collaboration.

Top Co-Authors

Avatar

Joseph D. Buxbaum

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Claudia Bagni

Katholieke Universiteit Leuven

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alexander Kolevzon

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Bernie Devlin

University of Pittsburgh

View shared research outputs
Top Co-Authors

Avatar

Hala Harony-Nicolas

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Arthur P. Goldberg

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Christopher S. Poultney

Icahn School of Medicine at Mount Sinai

View shared research outputs
Top Co-Authors

Avatar

Kathryn Roeder

Carnegie Mellon University

View shared research outputs
Top Co-Authors

Avatar

Lambertus Klei

University of Pittsburgh

View shared research outputs
Researchain Logo
Decentralizing Knowledge