Network


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

Hotspot


Dive into the research topics where Pietro Fratta is active.

Publication


Featured researches published by Pietro Fratta.


Science | 2014

C9orf72 repeat expansions cause neurodegeneration in Drosophila through arginine-rich proteins.

Sarah Mizielinska; Sebastian Grönke; Teresa Niccoli; Charlotte E. Ridler; Emma L. Clayton; Anny Devoy; Thomas Moens; Frances E. Norona; Ione O.C. Woollacott; Julian Pietrzyk; Karen Cleverley; Andrew J. Nicoll; Stuart Pickering-Brown; Jacqueline Dols; Melissa Cabecinha; Oliver Hendrich; Pietro Fratta; Elizabeth M. C. Fisher; Linda Partridge; Adrian M. Isaacs

Dipeptide repeat peptides on the attack Certain neurodegenerative diseases, including amyotrophic lateral sclerosis (ALS), are associated with expanded dipeptides translated from RNA transcripts of disease-associated genes (see the Perspective by West and Gitler). Kwon et al. show that the peptides encoded by the expanded repeats in the C9orf72 gene interfere with the way cells make RNA and kill cells. These effects may account for how this genetic form of ALS causes disease. Working in Drosophila, Mizielinska et al. aimed to distinguish between the effects of repeat-containing RNAs and the dipeptide repeat peptides that they encode. The findings provide evidence that dipeptide repeat proteins can cause toxicity directly. Science, this issue p. 1139 and p. 1192; see also p. 1118 In flies, arginine-rich proteins and RNA repeats contribute to a common genetic cause of neuronal cell death. [Also see Perspective by West and Gitler] An expanded GGGGCC repeat in C9orf72 is the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis. A fundamental question is whether toxicity is driven by the repeat RNA itself and/or by dipeptide repeat proteins generated by repeat-associated, non-ATG translation. To address this question, we developed in vitro and in vivo models to dissect repeat RNA and dipeptide repeat protein toxicity. Expression of pure repeats, but not stop codon–interrupted “RNA-only” repeats in Drosophila caused adult-onset neurodegeneration. Thus, expanded repeats promoted neurodegeneration through dipeptide repeat proteins. Expression of individual dipeptide repeat proteins with a non-GGGGCC RNA sequence revealed that both poly-(glycine-arginine) and poly-(proline-arginine) proteins caused neurodegeneration. These findings are consistent with a dual toxicity mechanism, whereby both arginine-rich proteins and repeat RNA contribute to C9orf72-mediated neurodegeneration.


American Journal of Human Genetics | 2013

Large C9orf72 Hexanucleotide Repeat Expansions Are Seen in Multiple Neurodegenerative Syndromes and Are More Frequent Than Expected in the UK Population

Jon Beck; Mark Poulter; Davina Hensman; Jonathan D. Rohrer; Colin J. Mahoney; Gary Adamson; Tracy Campbell; James Uphill; Aaron Borg; Pietro Fratta; Richard W. Orrell; Andrea Malaspina; James B. Rowe; Jeremy M Brown; John R. Hodges; Katie Sidle; James M. Polke; Henry Houlden; Jonathan M. Schott; Nick C. Fox; Sarah J. Tabrizi; Adrian M. Isaacs; John Hardy; Jason D. Warren; John Collinge; Simon Mead

Hexanucleotide repeat expansions in C9orf72 are a major cause of frontotemporal lobar degeneration (FTLD) and amyotrophic lateral sclerosis (ALS). Understanding the disease mechanisms and a method for clinical diagnostic genotyping have been hindered because of the difficulty in estimating the expansion size. We found 96 repeat-primed PCR expansions: 85/2,974 in six neurodegenerative diseases cohorts (FTLD, ALS, Alzheimer disease, sporadic Creutzfeldt-Jakob disease, Huntington disease-like syndrome, and other nonspecific neurodegenerative disease syndromes) and 11/7,579 (0.15%) in UK 1958 birth cohort (58BC) controls. With the use of a modified Southern blot method, the estimated expansion range (smear maxima) in cases was 800-4,400. Similarly, large expansions were detected in the population controls. Differences in expansion size and morphology were detected between DNA samples from tissue and cell lines. Of those in whom repeat-primed PCR detected expansions, 68/69 were confirmed by blotting, which was specific for greater than 275 repeats. We found that morphology in the expansion smear varied among different individuals and among different brain regions in the same individual. Expansion size correlated with age at clinical onset but did not differ between diagnostic groups. Evidence of instability of repeat size in control families, as well as neighboring SNP and microsatellite analyses, support multiple expansion events on the same haplotype background. Our method of estimating the size of large expansions has potential clinical utility. C9orf72-related disease might mimic several neurodegenerative disorders and, with potentially 90,000 carriers in the United Kingdom, is more common than previously realized.


Scientific Reports | 2012

C9orf72 hexanucleotide repeat associated with amyotrophic lateral sclerosis and frontotemporal dementia forms RNA G-quadruplexes

Pietro Fratta; Sarah Mizielinska; Andrew J. Nicoll; Mire Zloh; Elizabeth M. C. Fisher; Gary N. Parkinson; Adrian M. Isaacs

Large expansions of a non-coding GGGGCC-repeat in the first intron of the C9orf72 gene are a common cause of both amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). G-rich sequences have a propensity for forming highly stable quadruplex structures in both RNA and DNA termed G-quadruplexes. G-quadruplexes have been shown to be involved in a range of processes including telomere stability and RNA transcription, splicing, translation and transport. Here we show using NMR and CD spectroscopy that the C9orf72 hexanucleotide expansion can form a stable G-quadruplex, which has profound implications for disease mechanism in ALS and FTD.


Lancet Neurology | 2015

C9orf72 expansions in frontotemporal dementia and amyotrophic lateral sclerosis

Jonathan D. Rohrer; Adrian M. Isaacs; Sarah Mizielinska; Simon Mead; Tammaryn Lashley; Selina Wray; Katie Sidle; Pietro Fratta; Richard W. Orrell; John Hardy; Janice L. Holton; Tamas Revesz; Jason D. Warren

C9orf72 hexanucleotide repeat expansions are the most common cause of familial frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) worldwide. The clinical presentation is often indistinguishable from classic FTD or ALS, although neuropsychiatric symptoms are more prevalent and, for ALS, behavioural and cognitive symptoms occur more frequently. Pathogenic repeat length is in the hundreds or thousands, but the minimum length that increases risk of disease, and how or whether the repeat size affects phenotype, are unclear. Like in many patients with FTD and ALS, neuronal inclusions that contain TARDBP are seen, but are not universal, and the characteristic pathological finding is of dipeptide repeat (DPR) proteins, formed by unconventional repeat-associated non-ATG translation. Possible mechanisms of neurodegeneration include loss of C9orf72 protein and function, RNA toxicity, and toxicity from the DPR proteins, but which of these is the major pathogenic mechanism is not yet certain.


Neurology | 2015

Neurofilament light chain: A prognostic biomarker in amyotrophic lateral sclerosis.

Ching-Hua Lu; Corrie Macdonald-Wallis; Elizabeth Gray; Neil Pearce; Axel Petzold; Niklas Norgren; Gavin Giovannoni; Pietro Fratta; Katie Sidle; Mark Fish; Richard W. Orrell; Robin Howard; Kevin Talbot; Linda Greensmith; Jens Kuhle; Martin Turner; Andrea Malaspina

Objective: To test blood and CSF neurofilament light chain (NfL) levels in relation to disease progression and survival in amyotrophic lateral sclerosis (ALS). Methods: Using an electrochemiluminescence immunoassay, NfL levels were measured in samples from 2 cohorts of patients with sporadic ALS and healthy controls, recruited in London (ALS/control, plasma: n = 103/42) and Oxford (ALS/control, serum: n = 64/36; paired CSF: n = 38/20). NfL levels in patients were measured at regular intervals for up to 3 years. Change in ALS Functional Rating Scale–Revised score was used to assess disease progression. Survival was evaluated using Cox regression and Kaplan–Meier analysis. Results: CSF, serum, and plasma NfL discriminated patients with ALS from healthy controls with high sensitivity (97%, 89%, 90%, respectively) and specificity (95%, 75%, 71%, respectively). CSF NfL was highly correlated with serum levels (r = 0.78, p < 0.0001). Blood NfL levels were approximately 4 times as high in patients with ALS compared with controls in both cohorts, and maintained a relatively constant expression during follow-up. Blood NfL levels at recruitment were strong, independent predictors of survival. The highest tertile of blood NfL at baseline had a mortality hazard ratio of 3.91 (95% confidence interval 1.98–7.94, p < 0.001). Conclusion: Blood-derived NfL level is an easily accessible biomarker with prognostic value in ALS. The individually relatively stable levels longitudinally offer potential for NfL as a pharmacodynamic biomarker in future therapeutic trials. Classification of evidence: This report provides Class III evidence that the NfL electrochemiluminescence immunoassay accurately distinguishes patients with sporadic ALS from healthy controls.


Acta Neuropathologica | 2013

Homozygosity for the C9orf72 GGGGCC repeat expansion in frontotemporal dementia.

Pietro Fratta; Mark Poulter; Tammaryn Lashley; Jonathan D. Rohrer; James M. Polke; Jon Beck; Natalie S. Ryan; Davina Hensman; Sarah Mizielinska; Adrian James Waite; Mang-Ching Lai; Tania F. Gendron; Leonard Petrucelli; Elizabeth M. C. Fisher; Tamas Revesz; Jason D. Warren; John Collinge; Adrian M. Isaacs; Simon Mead

An expanded hexanucleotide repeat in the C9orf72 gene is the most common genetic cause of frontotemporal dementia and amyotrophic lateral sclerosis (c9FTD/ALS). We now report the first description of a homozygous patient and compare it to a series of heterozygous cases. The patient developed early-onset frontotemporal dementia without additional features. Neuropathological analysis showed c9FTD/ALS characteristics, with abundant p62-positive inclusions in the frontal and temporal cortices, hippocampus and cerebellum, as well as less abundant TDP-43-positive inclusions. Overall, the clinical and pathological features were severe, but did not fall outside the usual disease spectrum. Quantification of C9orf72 transcript levels in post-mortem brain demonstrated expression of all known C9orf72 transcript variants, but at a reduced level. The pathogenic mechanisms by which the hexanucleotide repeat expansion causes disease are unclear and both gain- and loss-of-function mechanisms may play a role. Our data support a gain-of-function mechanism as pure homozygous loss of function would be expected to lead to a more severe, or completely different clinical phenotype to the one described here, which falls within the usual range. Our findings have implications for genetic counselling, highlighting the need to use genetic tests that distinguish C9orf72 homozygosity.


Brain | 2013

Is SOD1 loss of function involved in amyotrophic lateral sclerosis

Rachele A. Saccon; Rosie K. A. Bunton-Stasyshyn; Elizabeth M. C. Fisher; Pietro Fratta

Mutations in the gene superoxide dismutase 1 (SOD1) are causative for familial forms of the neurodegenerative disease amyotrophic lateral sclerosis. When the first SOD1 mutations were identified they were postulated to give rise to amyotrophic lateral sclerosis through a loss of function mechanism, but experimental data soon showed that the disease arises from a—still unknown—toxic gain of function, and the possibility that loss of function plays a role in amyotrophic lateral sclerosis pathogenesis was abandoned. Although loss of function is not causative for amyotrophic lateral sclerosis, here we re-examine two decades of evidence regarding whether loss of function may play a modifying role in SOD1–amyotrophic lateral sclerosis. From analysing published data from patients with SOD1–amyotrophic lateral sclerosis, we find a marked loss of SOD1 enzyme activity arising from almost all mutations. We continue to examine functional data from all Sod1 knockout mice and we find obvious detrimental effects within the nervous system with, interestingly, some specificity for the motor system. Here, we bring together historical and recent experimental findings to conclude that there is a possibility that SOD1 loss of function may play a modifying role in amyotrophic lateral sclerosis. This likelihood has implications for some current therapies aimed at knocking down the level of mutant protein in patients with SOD1–amyotrophic lateral sclerosis. Finally, the wide-ranging phenotypes that result from loss of function indicate that SOD1 gene sequences should be screened in diseases other than amyotrophic lateral sclerosis.


Mammalian Genome | 2011

SOD1 and TDP-43 animal models of amyotrophic lateral sclerosis: recent advances in understanding disease toward the development of clinical treatments

Peter I. Joyce; Pietro Fratta; Elizabeth M. C. Fisher; Abraham Acevedo-Arozena

Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron disease with no cure. Breakthroughs in understanding ALS pathogenesis came with the discovery of dominant mutations in the superoxide dismutase 1 gene (SOD1) and other genes, including the gene encoding transactivating response element DNA binding protein-43 (TDP-43). This has led to the creation of animal models to further our understanding of the disease and identify a number of ALS-causing mechanisms, including mitochondrial dysfunction, protein misfolding and aggregation, oxidative damage, neuronal excitotoxicity, non-cell autonomous effects and neuroinflammation, axonal transport defects, neurotrophin depletion, effects from extracellular mutant SOD1, and aberrant RNA processing. Here we summarise the SOD1 and TDP-43 animal models created to date, report on recent findings supporting the potential mechanisms of ALS pathogenesis, and correlate this understanding with current developments in the clinic.


FEBS Letters | 2015

G-quadruplexes: Emerging roles in neurodegenerative diseases and the non-coding transcriptome

Roberto Simone; Pietro Fratta; Stephen Neidle; Gary N. Parkinson; Adrian M. Isaacs

G‐rich sequences in DNA and RNA have a propensity to fold into stable secondary structures termed G‐quadruplexes. G‐quadruplex forming sequences are widespread throughout the human genome, within both, protein coding and non‐coding genes, and regulatory regions. G‐quadruplexes have been implicated in multiple cellular functions including chromatin epigenetic regulation, DNA recombination, transcriptional regulation of gene promoters and enhancers, and translation. Here we will review the evidence for the occurrence of G‐quadruplexes both in vitro and in vivo; their role in neurological diseases including G‐quadruplex‐forming repeat expansions in the C9orf72 gene in frontotemporal dementia and amyotrophic lateral sclerosis and loss of the G‐quadruplex binding protein FMRP in the intellectual disability fragile X syndrome. We also review mounting evidence that supports a role for G‐quadruplexes in regulating the processing or function of a range of non‐coding RNAs. Finally we will highlight current perspectives for therapeutic interventions that target G‐quadruplexes.


Neurology | 2004

Mutant ubiquitin UBB+1 is accumulated in sporadic inclusion-body myositis muscle fibers

Pietro Fratta; Wk Engel; F. W. van Leeuwen; Elly M. Hol; G. Vattemi; V. Askanas

Mutant ubiquitin (UBB+1), a product of “molecular misreading,” is toxic to cells because its ubiquitinated form inhibits the proteasome, contributing to accumulation of misfolded proteins and their ensuing toxicity. The authors demonstrate in 10 sporadic inclusion body myositis (s-IBM) muscle biopsies that UBB+1 is accumulated in aggregates containing amyloid-β and phosphorylated-tau. In s-IBM, UBB+1 may be pathogenic by inhibiting proteasome, thereby promoting accumulation of cytotoxic misfolded amyloid-β and phosphorylated-tau.

Collaboration


Dive into the Pietro Fratta's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Linda Greensmith

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Adrian M. Isaacs

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Andrea Malaspina

Queen Mary University of London

View shared research outputs
Top Co-Authors

Avatar

Katie Sidle

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Michael G. Hanna

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Vincent Plagnol

University College London

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Hardy

University College London

View shared research outputs
Researchain Logo
Decentralizing Knowledge