Network


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

Hotspot


Dive into the research topics where Giovanna Zorzi is active.

Publication


Featured researches published by Giovanna Zorzi.


Nature Genetics | 2006

PLA2G6, encoding a phospholipase A2, is mutated in neurodegenerative disorders with high brain iron.

Neil V. Morgan; Shawn K. Westaway; Jenny Morton; Allison Gregory; Paul Gissen; Scott Sonek; Hakan Cangul; Jason Coryell; Natalie Canham; Nardo Nardocci; Giovanna Zorzi; Shanaz Pasha; Diana Rodriguez; Isabelle Desguerre; Amar Mubaidin; Enrico Bertini; Richard C. Trembath; Alessandro Simonati; Carolyn Schanen; Colin A. Johnson; Barbara Levinson; C. Geoffrey Woods; Beth Wilmot; Patricia L. Kramer; Jane Gitschier; Eamonn R. Maher; Susan J. Hayflick

Neurodegenerative disorders with high brain iron include Parkinson disease, Alzheimer disease and several childhood genetic disorders categorized as neuroaxonal dystrophies. We mapped a locus for infantile neuroaxonal dystrophy (INAD) and neurodegeneration with brain iron accumulation (NBIA) to chromosome 22q12-q13 and identified mutations in PLA2G6, encoding a calcium-independent group VI phospholipase A2, in NBIA, INAD and the related Karak syndrome. This discovery implicates phospholipases in the pathogenesis of neurodegenerative disorders with iron dyshomeostasis.


American Journal of Human Genetics | 2012

Exome sequencing reveals de novo WDR45 mutations causing a phenotypically distinct, X-linked dominant form of NBIA.

Tobias B. Haack; Penelope Hogarth; Michael C. Kruer; Allison Gregory; Thomas Wieland; Thomas Schwarzmayr; Elisabeth Graf; Lynn Sanford; Esther Meyer; Eleanna Kara; Stephan M. Cuno; Sami I. Harik; Vasuki H. Dandu; Nardo Nardocci; Giovanna Zorzi; Todd Dunaway; Mark A. Tarnopolsky; Steven Skinner; Steven J. Frucht; Era Hanspal; Connie Schrander-Stumpel; Delphine Héron; Cyril Mignot; Barbara Garavaglia; Kailash P. Bhatia; John Hardy; Tim M. Strom; Nathalie Boddaert; Henry Houlden; Manju A. Kurian

Neurodegeneration with brain iron accumulation (NBIA) is a group of genetic disorders characterized by abnormal iron deposition in the basal ganglia. We report that de novo mutations in WDR45, a gene located at Xp11.23 and encoding a beta-propeller scaffold protein with a putative role in autophagy, cause a distinctive NBIA phenotype. The clinical features include early-onset global developmental delay and further neurological deterioration (parkinsonism, dystonia, and dementia developing by early adulthood). Brain MRI revealed evidence of iron deposition in the substantia nigra and globus pallidus. Males and females are phenotypically similar, an observation that might be explained by somatic mosaicism in surviving males and germline or somatic mutations in females, as well as skewing of X chromosome inactivation. This clinically recognizable disorder is among the more common forms of NBIA, and we suggest that it be named accordingly as beta-propeller protein-associated neurodegeneration.


Neurology | 2008

Neurodegeneration associated with genetic defects in phospholipase A2

Allison Gregory; Shawn K. Westaway; I. E. Holm; Paul T. Kotzbauer; Penny Hogarth; Scott Sonek; J. C. Coryell; T. M. Nguyen; Nardo Nardocci; Giovanna Zorzi; D. Rodriguez; Isabelle Desguerre; Enrico Bertini; Alessandro Simonati; Barbara Levinson; Cristina Dias; Clara Barbot; Inês Carrilho; Manuela Santos; Ibrahim Malik; Jane Gitschier; Susan J. Hayflick

Objective: Mutations in the gene encoding phospholipase A2 group VI (PLA2G6) are associated with two childhood neurologic disorders: infantile neuroaxonal dystrophy (INAD) and idiopathic neurodegeneration with brain iron accumulation (NBIA). INAD is a severe progressive psychomotor disorder in which axonal spheroids are found in brain, spinal cord, and peripheral nerves. High globus pallidus iron is an inconsistent feature of INAD; however, it is a diagnostic criterion of NBIA, which describes a clinically and genetically heterogeneous group of disorders that share this hallmark feature. We sought to delineate the clinical, radiographic, pathologic, and genetic features of disease resulting from defective phospholipase A2. Methods: We identified 56 patients clinically diagnosed with INAD and 23 with idiopathic NBIA and screened their DNA for PLA2G6 mutations. Results: Eighty percent of patients with INAD had mutations in PLA2G6, whereas mutations were found in only 20% of those with idiopathic NBIA. All patients with two null mutations had a more severe phenotype. On MRI, nearly all mutation-positive patients had cerebellar atrophy, and half showed brain iron accumulation. We observed Lewy bodies and neurofibrillary tangles in association with PLA2G6 mutations. Conclusion: Defects in phospholipase A2 lead to a range of phenotypes. PLA2G6 mutations are associated with nearly all cases of classic infantile neuroaxonal dystrophy but a minority of cases of idiopathic neurodegeneration with brain iron accumulation, and genotype correlates with phenotype. Cerebellar atrophy predicts which patients are likely to be mutation-positive. The neuropathologic changes that are caused by defective phospholipase A2 suggest a shared pathogenesis with both Parkinson and Alzheimer diseases.


Annals of Neurology | 2006

Genotypic and phenotypic spectrum of PANK2 mutations in patients with neurodegeneration with brain iron accumulation

Monika Hartig; Konstanze Hörtnagel; Barbara Garavaglia; Giovanna Zorzi; Tomasz Kmiec; Thomas Klopstock; Kevin Rostasy; Marina Svetel; Vladimir Kostic; Markus Schuelke; Evelyn Botz; Adolf Weindl; Ivana Novakovic; Nardo Nardocci; Holger Prokisch; Thomas Meitinger

Neurodegeneration with brain iron accumulation (NBIA) is a group of disorders characterized by magnetic resonance imaging (MRI) changes in basal ganglia. Both missense and nonsense mutations have been found in such patients in a gene encoding the mitochondrial pantothenate kinase (PANK2).


Movement Disorders | 2005

Stimulation of the globus pallidus internus for childhood-onset dystonia

Giovanna Zorzi; Carlo Efisio Marras; Nardo Nardocci; Angelo Franzini; Luisa Chiapparini; Elio Maccagnano; Lucia Angelini; Dario Caldiroli; Giovanni Broggi

We report the results of deep brain stimulation (DBS) of the globus pallidus internus (GPi) in 12 patients with childhood‐onset generalized dystonia refractory to medication, including 3 patients with status dystonicus. There were 8 patients who had DYT1‐negative primary dystonia, 1 had DYT1‐positive dystonia, and 3 had symptomatic dystonia. Stimulation was effective in all but 1 patient. Dystonic postures and movements of the axis and limbs responded to DBS to a greater extent than oromandibular dystonia and fixed dystonic postures. These findings provide further evidence that pallidal stimulation is an effective treatment for intractable childhood‐onset dystonia, including status dystonicus, and together with previous findings, suggest that it should be considered the treatment of choice for these conditions.


Neurology | 1999

Infantile neuroaxonal dystrophy Clinical spectrum and diagnostic criteria

Nardo Nardocci; Giovanna Zorzi; Laura Farina; Simona Binelli; W. Scaioli; Claudia Ciano; L. Verga; Lucia Angelini; Mario Savoiardo; O. Bugiani

Objective: To present clinical, neurophysiologic, and neuroradiologic findings in 13 patients with infantile neuroaxonal dystrophy (INAD), focusing on aspects that assist early diagnosis. Background: Clinicopathologic diagnostic criteria for INAD were delineated by Aicardi and Castelein in 1979, but atypical cases are reported frequently and little is known of the diagnostic utility of MRI. Methods: The authors reviewed the clinical, neurophysiologic, and MRI findings of 13 patients who met the diagnostic criteria for INAD. Results: Symptoms onset was between 6 months and 2 years of age. In nine patients the clinical course was typical, with rapid motor and mental deterioration; in four patients progression was slower and the clinical picture was different. Electromyographic (EMG) signs of chronic denervation, fast rhythms on EEG and abnormal visual evoked potentials were observed in all patients during the disease course. Cerebellar atrophy with signal hyperintensity in the cerebellar cortex on T2-weighted images were the most characteristic MRI findings; hypointensity in the pallida and substantia nigra was also observed in two patients. α-N-acetyl-galactosaminidase activity on leukocytes was normal in the 10 patients tested. Conclusions: EMG and MRI abnormalities are the earliest and most suggestive signs of INAD, which has a clinical and radiologic spectrum that is broader than reported previously.


Neurological Sciences | 2004

Ataxia with isolated vitamin E deficiency: neurological phenotype, clinical follow-up and novel mutations in TTPAgene in Italian families

C. Mariotti; C. Gellera; M. Rimoldi; R. Mineri; Graziella Uziel; Giovanna Zorzi; Davide Pareyson; G. Piccolo; D. Gambi; Sylvie Piacentini; Ferdinando Squitieri; R. Capra; Barbara Castellotti; S. Di Donato

Abstract.Ataxia with vitamin E deficiency (AVED) is a rare autosomal recessive neurodegenerative disorder due to mutations in the alpha-tocopherol transfer protein (TTPA) gene on chromosome 8q13. AVED patients have progressive spinocerebellar symptoms and markedly reduced plasma levels of vitamin E. We studied neurological phenotype at diagnosis, and long-term effect of vitamin E supplementation in 16 patients from 12 Italian families. The most common mutations were the 744delA and 513insTT. Two novel TTPA mutations were identified: a severe truncating mutation (219insAT) in a homozygous patient, and a Gly246Arg missense mutation (G246R) in a compound heterozygous patient. The missense mutation was associated with a mild and slowly progressive form of the disease. Vitamin E supplementation therapy allowed a stabilization of the neurological conditions in most of the patients. However, development of spasticity and retinitis pigmentosa was noted in a few patients during therapy. Prompt genetic characterization of AVED patients may allow an effective early treatment and an adequate genetic counseling.


American Journal of Human Genetics | 2014

Exome sequence reveals mutations in CoA synthase as a cause of neurodegeneration with brain iron accumulation

Sabrina Dusi; Lorella Valletta; Tobias B. Haack; Yugo Tsuchiya; Paola Venco; Marco Tigano; Nikita Demchenko; Thomas Wieland; Thomas Schwarzmayr; Tim M. Strom; Federica Invernizzi; Barbara Garavaglia; Allison Gregory; Lynn Sanford; Jeffrey Hamada; Conceição Bettencourt; Henry Houlden; Luisa Chiapparini; Giovanna Zorzi; Manju A. Kurian; Nardo Nardocci; Holger Prokisch; Susan J. Hayflick; Ivan Gout; Valeria Tiranti

Neurodegeneration with brain iron accumulation (NBIA) comprises a clinically and genetically heterogeneous group of disorders with progressive extrapyramidal signs and neurological deterioration, characterized by iron accumulation in the basal ganglia. Exome sequencing revealed the presence of recessive missense mutations in COASY, encoding coenzyme A (CoA) synthase in one NBIA-affected subject. A second unrelated individual carrying mutations in COASY was identified by Sanger sequence analysis. CoA synthase is a bifunctional enzyme catalyzing the final steps of CoA biosynthesis by coupling phosphopantetheine with ATP to form dephospho-CoA and its subsequent phosphorylation to generate CoA. We demonstrate alterations in RNA and protein expression levels of CoA synthase, as well as CoA amount, in fibroblasts derived from the two clinical cases and in yeast. This is the second inborn error of coenzyme A biosynthesis to be implicated in NBIA.


Movement Disorders | 2011

Iron-related MRI images in patients with pantothenate kinase-associated neurodegeneration (PKAN) treated with deferiprone: Results of a phase II pilot trial

Giovanna Zorzi; Federica Zibordi; Luisa Chiapparini; Enrico Bertini; Lidia Russo; Antonio Piga; Filomena Longo; Barbara Garavaglia; Domenico Aquino; Mario Savoiardo; Alessandra Solari; Nardo Nardocci

The safety and efficacy of the oral iron‐chelating agent deferiprone on magnetic resonance pallida iron concentration and on clinical status were investigated in 10 patients affected by pantothenate kinase–associated neurodegeneration.


Molecular Genetics and Metabolism | 2012

Metabolic consequences of mitochondrial coenzyme A deficiency in patients with PANK2 mutations.

Valerio Leoni; Laura Strittmatter; Giovanna Zorzi; Federica Zibordi; Sabrina Dusi; Barbara Garavaglia; Paola Venco; Claudio Caccia; Amanda Souza; Amy Deik; Clary B. Clish; M. Rimoldi; Emilio Ciusani; Enrico Bertini; Nardo Nardocci; Vamsi K. Mootha; Valeria Tiranti

Pantothenate kinase-associated neurodegeneration (PKAN) is a rare, inborn error of metabolism characterized by iron accumulation in the basal ganglia and by the presence of dystonia, dysarthria, and retinal degeneration. Mutations in pantothenate kinase 2 (PANK2), the rate-limiting enzyme in mitochondrial coenzyme A biosynthesis, represent the most common genetic cause of this disorder. How mutations in this core metabolic enzyme give rise to such a broad clinical spectrum of pathology remains a mystery. To systematically explore its pathogenesis, we performed global metabolic profiling on plasma from a cohort of 14 genetically defined patients and 18 controls. Notably, lactate is elevated in PKAN patients, suggesting dysfunctional mitochondrial metabolism. As predicted, but never previously reported, pantothenate levels are higher in patients with premature stop mutations in PANK2. Global metabolic profiling and follow-up studies in patient-derived fibroblasts also reveal defects in bile acid conjugation and lipid metabolism, pathways that require coenzyme A. These findings raise a novel therapeutic hypothesis, namely, that dietary fats and bile acid supplements may hold potential as disease-modifying interventions. Our study illustrates the value of metabolic profiling as a tool for systematically exploring the biochemical basis of inherited metabolic diseases.

Collaboration


Dive into the Giovanna Zorzi's collaboration.

Top Co-Authors

Avatar

Nardo Nardocci

Carlo Besta Neurological Institute

View shared research outputs
Top Co-Authors

Avatar

Barbara Garavaglia

Carlo Besta Neurological Institute

View shared research outputs
Top Co-Authors

Avatar

Federica Zibordi

Carlo Besta Neurological Institute

View shared research outputs
Top Co-Authors

Avatar

Luisa Chiapparini

Carlo Besta Neurological Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Enrico Bertini

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Angelo Franzini

Catholic University of the Sacred Heart

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge