Network


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

Hotspot


Dive into the research topics where Pietro Spitali is active.

Publication


Featured researches published by Pietro Spitali.


Cell | 2012

Splice Modulating Therapies for Human Disease

Pietro Spitali; Annemieke Aartsma-Rus

Dysregulation of splicing and alternative splicing underlies many genetic and acquired diseases. We present an overview of recent strategies and successes in modulating splicing therapeutically in clinical and preclinical contexts. Effective approaches include restoring open reading frames, influencing alternative splicing, and inducing exon inclusion to generate beneficial proteins and remove deleterious ones.


Embo Molecular Medicine | 2014

Affinity proteomics within rare diseases: a BIO-NMD study for blood biomarkers of muscular dystrophies

Burcu Ayoglu; Amina Chaouch; Hanns Lochmüller; Luisa Politano; Enrico Bertini; Pietro Spitali; Monika Hiller; Eric H Niks; Francesca Gualandi; Fredrik Pontén; Kate Bushby; Annemieke Aartsma-Rus; Elena Schwartz; Yannick Le Priol; Volker Straub; Mathias Uhlén; Sebahattin Cirak; Peter A. C. 't Hoen; Francesco Muntoni; Alessandra Ferlini; Jochen M. Schwenk; Peter Nilsson; Cristina Al-Khalili Szigyarto

Despite the recent progress in the broad‐scaled analysis of proteins in body fluids, there is still a lack in protein profiling approaches for biomarkers of rare diseases. Scarcity of samples is the main obstacle hindering attempts to apply discovery driven protein profiling in rare diseases. We addressed this challenge by combining samples collected within the BIO‐NMD consortium from four geographically dispersed clinical sites to identify protein markers associated with muscular dystrophy using an antibody bead array platform with 384 antibodies. Based on concordance in statistical significance and confirmatory results obtained from analysis of both serum and plasma, we identified eleven proteins associated with muscular dystrophy, among which four proteins were elevated in blood from muscular dystrophy patients: carbonic anhydrase III (CA3) and myosin light chain 3 (MYL3), both specifically expressed in slow‐twitch muscle fibers and mitochondrial malate dehydrogenase 2 (MDH2) and electron transfer flavoprotein A (ETFA). Using age‐matched sub‐cohorts, 9 protein profiles correlating with disease progression and severity were identified, which hold promise for the development of new clinical tools for management of dystrophinopathies.


Proteomics Clinical Applications | 2014

Fibronectin is a serum biomarker for Duchenne muscular dystrophy

F. Cynthia Martin; Monika Hiller; Pietro Spitali; Stijn Oonk; Hans Dalebout; Magnus Palmblad; Amina Chaouch; M. Guglieri; Volker Straub; Hanns Lochmüller; Erik H. Niks; Jan J. Verschuuren; Annemieke Aartsma-Rus; André M. Deelder; Yuri E. M. van der Burgt; Peter A. C. 't Hoen

To identify and validate serum biomarkers for the progression of Duchenne muscular dystrophy (DMD) using a MS‐based bottom‐up pipeline.


Journal of Neurology, Neurosurgery, and Psychiatry | 2015

Validation of genetic modifiers for Duchenne muscular dystrophy: a multicentre study assessing SPP1 and LTBP4 variants

Janneke C. van den Bergen; Monika Hiller; Stefan Böhringer; Linda Vijfhuizen; H.B. Ginjaar; Amina Chaouch; Kate Bushby; Volker Straub; M. Scoto; Sebahattin Cirak; Véronique Humbertclaude; Mireille Claustres; C. Scotton; Chiara Passarelli; Hanns Lochmüller; Francesco Muntoni; Sylvie Tuffery-Giraud; Alessandra Ferlini; Annemieke Aartsma-Rus; Jan J. Verschuuren; Peter A. C. 't Hoen; Pietro Spitali

Objective Duchenne muscular dystrophy (DMD) is characterised by progressive muscle weakness. It has recently been reported that single nucleotide polymorphisms (SNPs) located in the SPP1 and LTBP4 loci can account for some of the inter-individual variability observed in the clinical disease course. The validation of genetic association in large independent cohorts is a key process for rare diseases in order to qualify prognostic biomarkers and stratify patients in clinical trials. Methods Duchenne patients from five European neuromuscular centres were included. Information about age at wheelchair dependence and steroid use was gathered. Melting curve analysis of PCR fragments or Sanger sequencing were used to genotype SNP rs28357094 in the SPP1 gene in 336 patients. The genotype of SNPs rs2303729, rs1131620, rs1051303 and rs10880 in the LTBP4 locus was determined in 265 patients by mass spectrometry. For both loci, a multivariate analysis was performed, using genotype/haplotype, steroid use and cohort as covariates. Results We show that corticosteroid treatment and the IAAM haplotype of the LTBP4 gene are significantly associated with prolonged ambulation in patients with DMD. There was no significant association between the SNP rs28357094 in the SPP1 gene and the age of ambulation loss. Conclusions This study underlines the importance of replicating genetic association studies for rare diseases in large independent cohorts to identify the most robust associations. We anticipate that genotyping of validated genetic associations will become important for the design and interpretation of clinical trials.


Laboratory Investigation | 2010

Accurate quantification of dystrophin mRNA and exon skipping levels in Duchenne Muscular Dystrophy

Pietro Spitali; Hans Heemskerk; Rolf H. A. M. Vossen; Alessandra Ferlini; Johan T. den Dunnen; Peter A. C. 't Hoen; Annemieke Aartsma-Rus

Antisense oligonucleotide (AON)-mediated exon skipping aimed at restoring the reading frame is a promising therapeutic approach for Duchenne muscular dystrophy that is currently tested in clinical trials. Numerous AONs have been tested in (patient-derived) cultured muscle cells and the mdx mouse model. The main outcome to measure AON efficiency is usually the exon-skipping percentage, though different groups use different methods to assess these percentages. Here, we compare a series of techniques to quantify exon skipping levels in AON-treated mdx mouse muscle. We compared densitometry of RT-PCR products on ethidium bromide-stained agarose gels, primary and nested RT-PCR followed by bioanalyzer analysis and melting curve analysis. The digital array system (Fluidigm) allows absolute quantification of skipped vs non-skipped transcripts and was used as a reference. Digital array results show that 1 ng of mdx gastrocnemius muscle-derived mRNA contains approximately 1100 dystrophin transcripts and that 665 transcripts are sufficient to determine exon-skipping levels. Quantification using bioanalyzer or densitometric analysis of primary PCR products resulted in values close to those obtained with digital array. The use of the same technique allows comparison between different groups working on exon skipping in the mdx mouse model.


Annals of Neurology | 2014

Reduced cerebral gray matter and altered white matter in boys with Duchenne muscular dystrophy.

N. Doorenweerd; C.S.M. Straathof; Eve M. Dumas; Pietro Spitali; Ieke B. Ginjaar; B.H.A. Wokke; D.G.M. Schrans; Janneke C. van den Bergen; Erik W. van Zwet; Andrew G. Webb; Mark A. van Buchem; Jan J. Verschuuren; Jos G.M. Hendriksen; Erik H. Niks; Hermien E. Kan

Duchenne muscular dystrophy (DMD) is characterized by progressive muscle weakness caused by DMD gene mutations leading to absence of the full‐length dystrophin protein in muscle. Multiple dystrophin isoforms are expressed in brain, but little is known about their function. DMD is associated with specific learning and behavioral disabilities that are more prominent in patients with mutations in the distal part of the DMD gene, predicted to affect expression of shorter protein isoforms. We used quantitative magnetic resonance (MR) imaging to study brain microstructure in DMD.


PLOS Currents | 2013

Autophagy is Impaired in the Tibialis Anterior of Dystrophin Null Mice

Pietro Spitali; Paolo Grumati; Monika Hiller; Martina Chrisam; Annemieke Aartsma-Rus; Paolo Bonaldo

Background Duchenne muscular dystrophy is a lethal, progressive, muscle-wasting disease caused by mutations in the DMD gene. Structural remodelling processes are responsible for muscle atrophy and replacement of myofibers by fibrotic and adipose tissues. Molecular interventions modulating catabolic pathways, such as the ubiquitin-proteasome and the autophagy-lysosome systems, are under development for Duchenne and other muscular dystrophies. The Akt signaling cascade is one of the main pathways involved in protein synthesis and autophagy repression and is known to be up-regulated in dystrophin null mdx mice. Results We report that autophagy is triggered by fasting in the tibialis anterior muscle of control mice but not in mdx mice. Mdx mice show persistent Akt activation upon fasting and failure to increase the expression of FoxO3 regulated autophagy and atrophy genes, such as Bnip3 and Atrogin1. We also provide evidence that autophagy is differentially regulated in mdx tibialis anterior and diaphragm muscles. Conclusions Our data support the concept that autophagy is impaired in the tibialis anterior muscle of mdx mice and that the regulation of autophagy is muscle type dependent. Differences between muscle groups should be considered during the pre-clinical development of therapeutic strategies addressing muscle metabolism.


The FASEB Journal | 2013

DMD transcript imbalance determines dystrophin levels

Pietro Spitali; Janneke C. van den Bergen; Ingrid E.C. Verhaart; B.H.A. Wokke; Anneke A.M. Janson; Rani van den Eijnde; Johan T. den Dunnen; Jeroen F. J. Laros; Jan J. Verschuuren; Peter A. C. 't Hoen; Annemieke Aartsma-Rus

Duchenne and Becker muscular dystrophies are caused by out‐of‐frame and in‐frame mutations, respectively, in the dystrophin encoding DMD gene. Molecular therapies targeting the precursormRNA are in clinical trials and show promising results. These approaches will depend on the stability and expression levels of dystrophin mRNA in skeletal muscles and heart. We report that the DMD gene is more highly expressed in heart than in skeletal muscles, in mice and humans. The transcript mutated in the mdx mouse model shows a 5′ to 3′ imbalance compared with that of its wild‐type counterpart and reading frame restoration via antisense‐mediated exon skipping does not correct this event. We also report significant transcript instability in 22 patients with Becker dystrophy, clarifying the fact that transcript imbalance is not caused by premature nonsense mutations. Finally, we demonstrate that transcript stability, rather than transcriptional rate, is an important determinant of dystrophin protein levels in patients with Becker dystrophy. We suggest that the availability of the complete transcript is a key factor to determine protein abundance and thus will influence the outcome of mRNA‐targeting therapies.—Spitali, P., van den Bergen, J. D., Verhaart, I. E. C., Wokke, B., Janson, A.A.M., van den Eijnde, R., den Dunnen, J. T., Laros, J. F. J., Verschuuren, J. G. M., t Hoen, P. A. C., Aartsma‐Rus, A., DMD transcript imbalance determines dystrophin levels. FASEB J. 27, 4909–4916 (2013). www.fasebj.org


Proteomics Clinical Applications | 2016

Comparative mass spectrometric and immunoassay-based proteome analysis in serum of Duchenne muscular dystrophy patients.

Stijn Oonk; Pietro Spitali; Monika Hiller; Linda Switzar; Hans Dalebout; Mattia Calissano; Hanns Lochmüller; Annemieke Aartsma-Rus; Peter A. C. 't Hoen; Yuri E. M. van der Burgt

Duchenne muscular dystrophy (DMD) is a severe and fatal neuromuscular disease. With the current developments on novel therapeutic strategies for DMD, the need to carefully monitor disease progression or regression upon treatment using molecular markers has become urgent.


Journal of neuromuscular diseases | 2015

Circulating Biomarkers for Duchenne Muscular Dystrophy

Annemieke Aartsma-Rus; Pietro Spitali

Abstract Duchenne muscular dystrophy is the most common form of muscular dystrophy. Genetic and biochemical research over the years has characterized the cause, pathophysiology and development of the disease providing several potential therapeutic targets and/or biomarkers. High throughput – omic technologies have provided a comprehensive understanding of the changes occurring in dystrophic muscles. Murine and canine animal models have been a valuable source to profile muscles and body fluids, thus providing candidate biomarkers that can be evaluated in patients. This review will illustrate known circulating biomarkers that could track disease progression and response to therapy in patients affected by Duchenne muscular dystrophy. We present an overview of the transcriptomic, proteomic, metabolomics and lipidomic biomarkers described in literature. We show how studies in muscle tissue have led to the identification of serum and urine biomarkers and we highlight the importance of evaluating biomarkers as possible surrogate endpoints to facilitate regulatory processes for new medicinal products.

Collaboration


Dive into the Pietro Spitali's collaboration.

Top Co-Authors

Avatar

Annemieke Aartsma-Rus

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Erik H. Niks

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Monika Hiller

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Peter A. C. 't Hoen

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Francesco Muntoni

Great Ormond Street Hospital

View shared research outputs
Top Co-Authors

Avatar

B.H.A. Wokke

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Hermien E. Kan

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Jan J. Verschuuren

Leiden University Medical Center

View shared research outputs
Top Co-Authors

Avatar

Andrew G. Webb

Leiden University Medical Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge