Network


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

Hotspot


Dive into the research topics where Simone Olgiati is active.

Publication


Featured researches published by Simone Olgiati.


Human Mutation | 2013

Mutation in the SYNJ1 gene associated with autosomal recessive, early-onset Parkinsonism.

Marialuisa Quadri; Mingyan Fang; Marina Picillo; Simone Olgiati; Guido J. Breedveld; Josja Graafland; Bin Wu; Fengping Xu; Roberto Erro; Marianna Amboni; Sabina Pappatà; Mario Quarantelli; Grazia Annesi; Aldo Quattrone; Hsin F. Chien; Egberto Reis Barbosa; Ben A. Oostra; Paolo Barone; Jun Wang; Vincenzo Bonifati

Autosomal recessive, early‐onset Parkinsonism is clinically and genetically heterogeneous. Here, we report the identification, by homozygosity mapping and exome sequencing, of a SYNJ1 homozygous mutation (p.Arg258Gln) segregating with disease in an Italian consanguineous family with Parkinsonism, dystonia, and cognitive deterioration. Response to levodopa was poor, and limited by side effects. Neuroimaging revealed brain atrophy, nigrostriatal dopaminergic defects, and cerebral hypometabolism. SYNJ1 encodes synaptojanin 1, a phosphoinositide phosphatase protein with essential roles in the postendocytic recycling of synaptic vesicles. The mutation is absent in variation databases and in ethnically matched controls, is damaging according to all prediction programs, and replaces an amino acid that is extremely conserved in the synaptojanin 1 homologues and in SAC1‐like domains of other proteins. Sequencing the SYNJ1 ORF in unrelated patients revealed another heterozygous mutation (p.Ser1422Arg), predicted as damaging, in a patient who also carries a heterozygous PINK1 truncating mutation. The SYNJ1 gene is a compelling candidate for Parkinsonism; mutations in the functionally linked protein auxilin cause a similar early‐onset phenotype, and other findings implicate endosomal dysfunctions in the pathogenesis. Our data delineate a novel form of human Mendelian Parkinsonism, and provide further evidence for abnormal synaptic vesicle recycling as a central theme in the pathogenesis.


Neurogenetics | 2014

PARK20 caused by SYNJ1 homozygous Arg258Gln mutation in a new Italian family

Simone Olgiati; Anna De Rosa; Marialuisa Quadri; Chiara Criscuolo; Guido J. Breedveld; Marina Picillo; Sabina Pappatà; Mario Quarantelli; Paolo Barone; Giuseppe De Michele; Vincenzo Bonifati

SYNJ1 has been recently identified by two independent groups as the gene defective in a novel form of autosomal recessive, early-onset atypical parkinsonism (PARK20). Two consanguineous families were initially reported (one of Sicilian and one of Iranian origins), with the same SYNJ1 homozygous mutation (c.773G > A, p.Arg258Gln) segregating with a similar phenotype of early-onset parkinsonism and additional atypical features. Here, we report the identification of the same SYNJ1 homozygous mutation in two affected siblings of a third pedigree. Both siblings had mild developmental psychomotor delay, followed, during the third decade of life, by progressive parkinsonism, dystonia, and mild cognitive impairment. One sibling suffered one episode of generalized seizures. Neuroimaging studies revealed severe nigrostriatal dopaminergic defects, mild striatal and very mild cortical hypometabolism. Treatment with dopamine agonists and anticholinergics resulted in partial improvements. Genetic analyses revealed in both siblings the SYNJ1 homozygous c.773G > A (p.Arg258Gln) mutation. Haplotype analysis suggests that the mutation has arisen independently in this family and the Sicilian PARK20 family previously described by us, in keeping with the hypothesis of a mutational hot spot. This is the third reported family with autosomal recessive, early-onset parkinsonism associated with the SYNJ1 p.Arg258Gln mutation. This work contributes to the definition of the genetic and clinical aspects of PARK20. This newly recognized form must be considered in the diagnostic work-up of patients with early-onset atypical parkinsonism. The presence of seizures might represent a red flag to suspect PARK20.


Annals of Neurology | 2016

DNAJC6 Mutations Associated with Early-Onset Parkinson's Disease

Simone Olgiati; Marialuisa Quadri; Mingyan Fang; Janneke P.M.A. Rood; Jonas Alex Morales Saute; Hsin Fen Chien; Christian G. Bouwkamp; Josja Graafland; Michelle Minneboo; Guido J. Breedveld; Jianguo Zhang; Frans W. Verheijen; Agnita J.W. Boon; Anneke J.A. Kievit; Laura Bannach Jardim; Wim Mandemakers; Egberto Reis Barbosa; Carlos Roberto de Mello Rieder; Klaus L. Leenders; Jun Wang; Vincenzo Bonifati

DNAJC6 mutations were recently described in two families with autosomal recessive juvenile parkinsonism (onset age < 11), prominent atypical signs, poor or absent response to levodopa, and rapid progression (wheelchair‐bound within ∼10 years from onset). Here, for the first time, we report DNAJC6 mutations in early‐onset Parkinsons disease (PD).


Parkinsonism & Related Disorders | 2014

Beta-propeller protein-associated neurodegeneration (BPAN), a rare form of NBIA: Novel mutations and neuropsychiatric phenotype in three adult patients

Willem M.A. Verhoeven; J.I.M. Egger; David A. Koolen; Helger G. Yntema; Simone Olgiati; Guido J. Breedveld; Vincenzo Bonifati; Bart P. van de Warrenburg

Neurodegeneration with brain iron accumulation (NBIA) comprises a group of rare neuropsychiatric syndromes characterized by iron accumulation in the basal ganglia. The pantothenate kinase-associated neurodegeneration (PKAN) was the first NBIA form to be genetically identified almost 15 years ago. Nowadays, eight types can be genetically distinguished. More recently, a novel NBIA was delineated and termed Static Encephalopathy of childhood with Neurodegeneration in Adulthood (SENDA), characterized by early intellectual disability followed by delayed progressive motor and cognitive deterioration with an onset in the second to third decade. Very recently, mutations in the WD repeat-containing protein 45 (WDR45) gene located on Xp11.23 were shown to be the causal factor. The protein encoded by WDR45 propels protein interaction important for autophagy. This form was therefore retermed Beta-propeller Protein Associated Neurodegeneration (BPAN). Here, the first three Dutch patients with genetically proven BPAN are comprehensively described with respect to course and neurological as well as neuropsychiatric phenotypes. All three showed a characteristic delayed progression of neurological symptoms with parkinsonism and prominent dystonia. Treatment with levodopa/carbidopa had limited effects only. Neuropsychiatric symptoms within the autistic and affective spectrum were present in the early phase of the disease. The specific course and prognosis should implicate restrained psychopharmacological interventions. The clinical picture and imaging hallmarks are often highly suggestive and should lead to suspect this specific disorder. However, the identification of a WDR45 mutation is needed for a definite diagnosis of BPAN.


Movement Disorders | 2014

Primary familial brain calcification: Genetic analysis and clinical spectrum

Ilaria Taglia; Andrea Mignarri; Simone Olgiati; Elisabetta Menci; Patrizia L. Petrocelli; Guido J. Breedveld; Cesa Scaglione; Paolo Martinelli; Antonio Federico; Vincenzo Bonifati; Maria Teresa Dotti

Primary familial brain calcification (PFBC) is a rare autosomal dominant disorder with bilateral calcification of basal ganglia and other cerebral regions, movement disorders, and neuropsychiatric disturbances. So far, three causative genes have been discovered: SLC20A2, PDGFRB and PDGFB, accounting for approximately 50% of cases.


Movement Disorders | 2015

Manganese Transport Disorder: Novel SLC30A10 Mutations and Early Phenotypes

Marialuisa Quadri; Dm Mahesh Kamate Md; Dm Suvasini Sharma Md; Simone Olgiati; Josja Graafland; Guido J. Breedveld; Indu Kori; Virupaxi Hattiholi; Dm Puneet Jain Md; Satinder Aneja; Atin Kumar; Parveen Gulati; Medha Goel; Bibek Talukdar; Vincenzo Bonifati

SLC30A10 mutations cause an autosomal recessive disorder, characterized by hypermanganesaemia, polycythemia, early‐onset dystonia, paraparesis, or late‐onset parkinsonism, and chronic liver disease. This is the first identified inborn error of Mn metabolism in humans, reported in 10 families thus far.


Parkinsonism & Related Disorders | 2015

Novel GCH1 variant in Dopa-responsive dystonia and Parkinson's disease

A.J. Lewthwaite; Td Lambert; E.B. Rolfe; Simone Olgiati; Marialuisa Quadri; Erik J. Simons; Karen E. Morrison; Vincenzo Bonifati; David Nicholl

Background GTP cyclohydrolase I (GCH1) mutations are the commonest cause of Dopa-responsive dystonia (DRD). Clinical phenotypes can be broad, even within a single family. Methods We present clinical, genetic and functional imaging data on a British kindred in which affected subjects display phenotypes ranging from DRD to Parkinsons disease (PD). Twelve family members were studied. Clinical examination, dopamine transporter (DAT) imaging, and molecular genetic analysis of GCH1 and the commonest known familial PD-related genes were performed. Results We have identified a novel missense variant, c.5A > G, p.(Glu2Gly), within the GCH1 gene in affected family members displaying a range of phenotypes. Two affected subjects carrying this variant had abnormal DAT imaging. These two with abnormal DAT imaging had a PD phenotype, while the remaining three subjects with the novel GCH1 variant had normal DAT imaging and a DRD phenotype. Conclusions We propose that this GCH1 variant is pathogenic in this family and these findings suggest that similar mechanisms involving abnormal GTP cyclohydolase I may underlie both PD and DRD. GCH1 genetic testing should be considered in patients with PD and a family history of DRD.


Movement Disorders | 2016

Genetics of movement disorders in the next-generation sequencing era

Simone Olgiati; Marialuisa Quadri; Vincenzo Bonifati

Several innovative and extremely powerful methods for sequencing nucleic acids (DNA and RNA), collectively known as next‐generation sequencing technologies, have become available in the past few years. The application of these technologies is rapidly changing the landscape of both medical genetic research and clinical practice: the pace of discovery of novel disease‐causing or disease‐predisposing genes is markedly accelerating; the phenotypic spectra associated with previously known genes is expanding; and novel tools for rapid, cheap, and comprehensive genetic testing are entering the clinical practice. As with every technological revolution, next‐generation sequencing also comes with new challenges concerning the storage, the analysis, and crucially, the interpretation of the large amounts of generated data. The current possibility to sequence entire human exomes (the coding part of the genome) or entire genomes at affordable costs has brought the era of personalized medicine closer than ever, also raising new legal and ethical issues. In this article, we summarize the essential technological aspects of next‐generation sequencing and discuss their applications in the field of movement disorders. We review the different strategies for gene finding enabled by these technologies (including project designs, filtering approaches, and bioinformatic tools) and we then discuss their applications in clinical practice.


Movement Disorders | 2016

Paroxysmal exercise-induced dystonia within the phenotypic spectrum of ECHS1 deficiency

Simone Olgiati; Matej Skorvanek; Marialuisa Quadri; Michelle Minneboo; Josja Graafland; Guido J. Breedveld; Ramon Bonte; Zeliha Ozgur; Mirjam C. G. N. van den Hout; Kees Schoonderwoerd; Frans W. Verheijen; Wilfred van IJcken; Hsin Fen Chien; Egberto Reis Barbosa; Hsiu Chen Chang; Szu Chia Lai; Tu Hsueh Yeh; Chin Song Lu; Yah Huei Wu-Chou; Anneke J.A. Kievit; Vladimir Han; Zuzana Gdovinova; Robert Jech; Robert M.W. Hofstra; George J. G. Ruijter; Wim Mandemakers; Vincenzo Bonifati

ECHS1 encodes a mitochondrial enzyme involved in the degradation of essential amino acids and fatty acids. Recently, ECHS1 mutations were shown to cause a new severe metabolic disorder presenting as Leigh or Leigh‐like syndromes. The objective of this study was to describe a family with 2 siblings affected by different dystonic disorders as a resulting phenotype of ECHS1 mutations.


Neurogenetics | 2015

An exome study of Parkinson's disease in Sardinia, a Mediterranean genetic isolate

Marialuisa Quadri; Xu Yang; Giovanni Cossu; Simone Olgiati; Valeria M. Saddi; Guido J. Breedveld; Limei Ouyang; Jingchu Hu; Na Xu; Josja Graafland; Valeria Ricchi; Daniela Murgia; Leonor Correia Guedes; Claudio Mariani; María José Martí; Patrizia Tarantino; Rosanna Asselta; Francesc Valldeoriola; Monica Gagliardi; Gianni Pezzoli; Mario Ezquerra; Aldo Quattrone; Joaquim Ferreira; Grazia Annesi; Stefano Goldwurm; Eduardo Tolosa; Ben A. Oostra; Maurizio Melis; Jun Wang; Vincenzo Bonifati

Parkinson’s disease (PD) is a common neurodegenerative disorder of complex aetiology. Rare, highly penetrant PD-causing mutations and common risk factors of small effect size have been identified in several genes/loci. However, these mutations and risk factors only explain a fraction of the disease burden, suggesting that additional, substantial genetic determinants remain to be found. Genetically isolated populations offer advantages for dissecting the genetic architecture of complex disorders, such as PD. We performed exome sequencing in 100 unrelated PD patients from Sardinia, a genetic isolate. SNPs absent from dbSNP129 and 1000 Genomes, shared by at least five patients, and of functional effects were genotyped in an independent Sardinian case-control sample (n = 500). Variants associated with PD with nominal p value <0.05 and those with odds ratio (OR) ≥3 were validated by Sanger sequencing and typed in a replication sample of 2965 patients and 2678 controls from Italy, Spain, and Portugal. We identified novel moderately rare variants in several genes, including SCAPER, HYDIN, UBE2H, EZR, MMRN2 and OGFOD1 that were specifically present in PD patients or enriched among them, nominating these as novel candidate risk genes for PD, although no variants achieved genome-wide significance after Bonferroni correction. Our results suggest that the genetic bases of PD are highly heterogeneous, with implications for the design of future large-scale exome or whole-genome analyses of this disease.

Collaboration


Dive into the Simone Olgiati's collaboration.

Top Co-Authors

Avatar

Marialuisa Quadri

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Vincenzo Bonifati

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Guido J. Breedveld

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Josja Graafland

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Wim Mandemakers

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Frans W. Verheijen

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar

Michelle Minneboo

Erasmus University Rotterdam

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Hsin Fen Chien

University of São Paulo

View shared research outputs
Top Co-Authors

Avatar

Jun Wang

Chinese Academy of Sciences

View shared research outputs
Researchain Logo
Decentralizing Knowledge