Network


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

Hotspot


Dive into the research topics where Emanuele Buratti is active.

Publication


Featured researches published by Emanuele Buratti.


Science | 2008

TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis

Jemeen Sreedharan; Ian P. Blair; Vineeta Tripathi; Xun Hu; Caroline Vance; Boris Rogelj; Steven Ackerley; Jennifer C Durnall; Kelly L. Williams; Emanuele Buratti; Francisco E. Baralle; Jacqueline de Belleroche; J. Douglas Mitchell; P. Nigel Leigh; Ammar Al-Chalabi; Christopher Miller; Garth A. Nicholson; Christopher Shaw

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disorder characterized pathologically by ubiquitinated TAR DNA binding protein (TDP-43) inclusions. The function of TDP-43 in the nervous system is uncertain, and a mechanistic role in neurodegeneration remains speculative. We identified neighboring mutations in a highly conserved region of TARDBP in sporadic and familial ALS cases. TARDBPM337V segregated with disease within one kindred and a genome-wide scan confirmed that linkage was restricted to chromosome 1p36, which contains the TARDBP locus. Mutant forms of TDP-43 fragmented in vitro more readily than wild type and, in vivo, caused neural apoptosis and developmental delay in the chick embryo. Our evidence suggests a pathophysiological link between TDP-43 and ALS.


The EMBO Journal | 2001

Nuclear factor TDP-43 and SR proteins promote in vitro and in vivo CFTR exon 9 skipping

Emanuele Buratti; Thilo Dörk; Elisabetta Zuccato; Franco Pagani; Maurizio Romano; Francisco E. Baralle

Alternative splicing of human cystic fibrosis transmembrane conductance regulator (CFTR) exon 9 is regulated by a combination of cis‐acting elements distributed through the exon and both flanking introns (IVS8 and IVS9). Several studies have identified in the IVS8 intron 3′ splice site a regulatory element that is composed of a polymorphic (TG)m(T)n repeated sequence. At present, no cellular factors have been identified that recognize this element. We have identified TDP‐43, a nuclear protein not previously described to bind RNA, as the factor binding specifically to the (TG)m sequence. Transient TDP‐43 overexpression in Hep3B cells results in an increase in exon 9 skipping. This effect is more pronounced with concomitant overexpression of SR proteins. Antisense inhibition of endogenous TDP‐43 expression results in increased inclusion of exon 9, providing a new therapeutic target to correct aberrant splicing of exon 9 in CF patients. The clinical and biological relevance of this finding in vivo is demonstrated by our characterization of a CF patient carrying a TG10T9(ΔF508)/TG13T3(wt) genotype leading to a disease‐causing high proportion of exon 9 skipping.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Aberrant cleavage of TDP-43 enhances aggregation and cellular toxicity

Yong Jie Zhang; Ya Fei Xu; Casey Cook; Tania F. Gendron; Paul S. Roettges; Christopher D. Link; Wen Lang Lin; Jimei Tong; Monica Castanedes-Casey; Peter E.A. Ash; Jennifer Gass; Vijayaraghavan Rangachari; Emanuele Buratti; Francisco E. Baralle; Todd E. Golde; Dennis W. Dickson; Leonard Petrucelli

Inclusions of TAR DNA-binding protein-43 (TDP-43), a nuclear protein that regulates transcription and RNA splicing, are the defining histopathological feature of frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTLD-Us) and sporadic and familial forms of amyotrophic lateral sclerosis (ALS). In ALS and FTLD-U, aggregated, ubiquitinated, and N-terminally truncated TDP-43 can be isolated from brain tissue rich in neuronal and glial cytoplasmic inclusions. The loss of TDP-43 function resulting from inappropriate cleavage, translocation from the nucleus, or its sequestration into inclusions could play important roles in neurodegeneration. However, it is not known whether TDP-43 fragments directly mediate toxicity and, more specifically, whether their abnormal aggregation is a cause or consequence of pathogenesis. We report that the ectopic expression of a ≈25-kDa TDP-43 fragment corresponding to the C-terminal truncation product of caspase-cleaved TDP-43 leads to the formation of toxic, insoluble, and ubiquitin- and phospho-positive cytoplasmic inclusions within cells. The 25-kDa C-terminal fragment is more prone to phosphorylation at S409/S410 than full-length TDP-43, but phosphorylation at these sites is not required for inclusion formation or toxicity. Although this fragment shows no biological activity, its exogenous expression neither inhibits the function nor causes the sequestration of full-length nuclear TDP-43, suggesting that the 25-kDa fragment can induce cell death through a toxic gain-of-function. Finally, by generating a conformation-dependent antibody that detects C-terminal fragments, we show that this toxic cleavage product is specific for pathologic inclusions in human TDP-43 proteinopathies.


Annals of Neurology | 2008

Phosphorylated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis.

Masato Hasegawa; Tetsuaki Arai; Takashi Nonaka; Fuyuki Kametani; Mari Yoshida; Yoshio Hashizume; Thomas G. Beach; Emanuele Buratti; Francisco E. Baralle; Mitsuya Morita; Imaharu Nakano; Tatsuro Oda; Kuniaki Tsuchiya; Haruhiko Akiyama

TAR DNA‐binding protein of 43kDa (TDP‐43) is deposited as cytoplasmic and intranuclear inclusions in brains of patients with frontotemporal lobar degeneration with ubiquitinated inclusions (FTLD‐U) and amyotrophic lateral sclerosis (ALS). Previous studies reported that abnormal phosphorylation takes place in deposited TDP‐43. The aim of this study was to identify the phosphorylation sites and responsible kinases, and to clarify the pathological significance of phosphorylation of TDP‐43.


Journal of Biological Chemistry | 2005

TDP-43 binds heterogeneous nuclear ribonucleoprotein A/B through its C-terminal tail : An important region for the inhibition of cystic fibrosis transmembrane conductance regulator exon 9 splicing

Emanuele Buratti; Antonia Brindisi; Maurizio Giombi; Sergio Tisminetzky; Youhna M. Ayala; Francisco E. Baralle

TDP-43 is a highly conserved nuclear factor of yet unknown function that binds to ug-repeated sequences and is responsible for cystic fibrosis transmembrane conductance regulator exon 9 splicing inhibition. We have analyzed TDP-43 interactions with other splicing factors and identified the critical regions for the protein/protein recognition events that determine this biological function. We show here that the C-terminal region of TDP-43 is capable of binding directly to several proteins of the heterogeneous nuclear ribonucleoprotein (hnRNP) family with well known splicing inhibitory activity, in particular, hnRNP A2/B1 and hnRNP A1. Mutational analysis showed that TDP-43 proteins lacking the C-terminal region could not inhibit splicing probably because they were unable to form the hnRNP-rich complex involved in splicing inhibition. Finally, through splicing complex analysis, we show that splicing inhibition mediated by TDP-43 occurs at the earliest stages of spliceosomal assembly.


Frontiers in Bioscience | 2008

Multiple roles of TDP-43 in gene expression, splicing regulation, and human disease

Emanuele Buratti; Francisco E. Baralle

TDP-43 is a RNA/DNA binding protein that structurally resembles a typical hnRNP protein family member and displays a significant specificity for binding the common microsatellite region (GU/GT)n. Initially described as a regulator of HIV-1 gene expression, it has been reported in the past to affect both normal and pathological RNA splicing events. In particular, it has been shown to play a fundamental role in the occurrence of several monosymptomatic/full forms of Cystic Fibrosis caused by pathological skipping of CFTR exon 9 from the mature mRNA. Recently, and in a way probably unrelated to splicing, a hyperphosphorylated form of TDP-43 has also been found to accumulate in the cytoplasm of neuronal cells of patients affected by fronto temporal lobar degenerations. In addition to its role in transcription and splicing regulation, a growing body of evidence indirectly suggests that TDP-43 may be involved in other cellular processes such as microRNA biogenesis, apoptosis, and cell division. The aim of this work is to provide the basic facts about TDP-43 an assessment of the multiple functions ascribed to this protein.


Molecular and Cellular Biology | 2004

Influence of RNA secondary structure on the pre-mRNA splicing process.

Emanuele Buratti; Francisco E. Baralle

Pre-mRNA splicing in eukaryotes requires joining together the nucleotides of the various mRNA-coding regions (exons) after recognizing them from the normally vastly superior number of non-mRNA-coding sequences (introns). For three excellent reviews on general splicing and its regulation, refer to references 14, 62, and 70. In eukaryotes, the vast majority of splicing processes are catalyzed by the spliceosome, a very complex RNA-protein aggregate which has been estimated to contain several hundred different proteins in addition to five spliceosomal snRNAs (1, 54, 62, 63, 81, 109). These factors are responsible for the accurate positioning of the spliceosome on the 5′ and 3′ splice site sequences. The reason why so many factors are needed reflects the observation that exon recognition can be affected by many pre-mRNA features such as exon length (5, 97), the presence of enhancer and silencer elements (8, 62), the strength of splicing signals (45), the promoter architecture (29, 55), and the rate of RNA processivity (86). In addition, the general cellular environment also exerts an effect, as recent observations suggest the existence of extensive coupling between splicing and many other gene expression steps (69) and even its modification by external stimuli (96).


The Journal of Neuroscience | 2007

Progranulin Mediates Caspase-Dependent Cleavage of TAR DNA Binding Protein-43

Yong Jie Zhang; Ya Fei Xu; Chad A. Dickey; Emanuele Buratti; Francisco E. Baralle; Rachel M. Bailey; Stuart Pickering-Brown; Dennis W. Dickson; Leonard Petrucelli

TAR DNA binding protein-43 (TDP-43) is the pathologic substrate of neuronal and glial inclusions in frontotemporal lobar degeneration with ubiquitin-positive inclusions (FTDL-U) and in amyotrophic lateral sclerosis (ALS). Mutations in the progranulin gene (PGRN) have been shown to cause familial FTLD-U. The relationship between progranulin and TDP-43 and their respective roles in neurodegeneration is unknown. We report that progranulin mediates proteolytic cleavage of TDP-43 to generate ∼35 and ∼25 kDa species. Suppression of PGRN expression with small interfering RNA leads to caspase-dependent accumulation of TDP-43 fragments that can be inhibited with caspase inhibitor treatment. Cells treated with staurosporine also induced caspase-dependent cleavage and redistribution of TDP-43 from its nuclear localization to cytoplasm. Altered cleavage and redistribution of TDP-43 in cell culture models are similar to findings in FTLD-U and ALS. The results suggest that abnormal metabolism of TDP-43 mediated by progranulin may play a pivotal role in neurodegeneration.


Journal of Cell Science | 2008

Structural determinants of the cellular localization and shuttling of TDP-43

Youhna M. Ayala; Paola Zago; Andrea D'Ambrogio; Yafei Xu; Leonard Petrucelli; Emanuele Buratti; Francisco E. Baralle

TDP-43 (also known as TARDBP) regulates different processes of gene expression, including transcription and splicing, through RNA and DNA binding. Moreover, recent reports have shown that the protein interacts with the 3′UTRs of specific mRNAs. The aberrant cellular distribution and aggregation of TDP-43 were recently reported in neurodegenerative diseases, namely frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). A detailed description of the determinants for cellular localization has yet to emerge, including information on how the known functions of TDP-43 and cellular targeting affect each other. We provide the first experimental evidence that TDP-43 continuously shuttles between nucleus and cytoplasm in a transcription-dependent manner. Furthermore, we investigate the role of the functional TDP-43 domains in determining cellular targeting through a combination of immunofluorescence and biochemical fractionation methods. Our analyses indicate that the C-terminus is essential for solubility and cellular localization, because its deletion results in the formation of large nuclear and cytoplasmic aggregates. Disruption of the RNA-recognition domain required for RNA and DNA binding, however, alters nuclear distribution by decreasing TDP-43 presence in the nucleoplasm. Our findings suggest that TDP-43 solubility and localization are particularly sensitive to disruptions that extend beyond the newly found nuclear localization signal and depend on a combination of factors that are closely connected to the functional properties of this protein.


The EMBO Journal | 2011

TDP‐43 regulates its mRNA levels through a negative feedback loop

Youhna M. Ayala; Laura De Conti; S. Eréndira Avendaño-Vázquez; Ashish Dhir; Maurizio Romano; Andrea D'Ambrogio; James Tollervey; Jernej Ule; Marco Baralle; Emanuele Buratti; Francisco E. Baralle

TAR DNA‐binding protein (TDP‐43) is an evolutionarily conserved heterogeneous nuclear ribonucleoprotein (hnRNP) involved in RNA processing, whose abnormal cellular distribution and post‐translational modification are key markers of certain neurodegenerative diseases, such as amyotrophic lateral sclerosis and frontotemporal lobar degeneration. We generated human cell lines expressing tagged forms of wild‐type and mutant TDP‐43 and observed that TDP‐43 controls its own expression through a negative feedback loop. The RNA‐binding properties of TDP‐43 are essential for the autoregulatory activity through binding to 3′ UTR sequences in its own mRNA. Our analysis indicated that the C‐terminal region of TDP‐43, which mediates TDP‐43–hnRNP interactions, is also required for self‐regulation. TDP‐43 binding to its 3′ UTR does not significantly change the pre‐mRNA splicing pattern but promotes RNA instability. Moreover, blocking exosome‐mediated degradation partially recovers TDP‐43 levels. Our findings demonstrate that cellular TDP‐43 levels are under tight control and it is likely that disease‐associated TDP‐43 aggregates disrupt TDP‐43 self‐regulation, thus contributing to pathogenesis.

Collaboration


Dive into the Emanuele Buratti's collaboration.

Top Co-Authors

Avatar

Francisco E. Baralle

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Cristiana Stuani

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Marco Baralle

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Sergio Tisminetzky

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Diana Baralle

University of Southampton

View shared research outputs
Top Co-Authors

Avatar

F.E. Baralle

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Franco Pagani

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Laura De Conti

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Top Co-Authors

Avatar

Youhna M. Ayala

International Centre for Genetic Engineering and Biotechnology

View shared research outputs
Researchain Logo
Decentralizing Knowledge