Network


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

Hotspot


Dive into the research topics where Marcello Niceta is active.

Publication


Featured researches published by Marcello Niceta.


American Journal of Human Genetics | 2012

A restricted spectrum of mutations in the SMAD4 tumor-suppressor gene underlies myhre syndrome

Viviana Caputo; Luciano Cianetti; Marcello Niceta; Claudio Carta; Andrea Ciolfi; Gianfranco Bocchinfuso; Eugenio Carrani; Maria Lisa Dentici; Elisa Biamino; E Belligni; Livia Garavelli; Loredana Boccone; Daniela Melis; Generoso Andria; Bruce D. Gelb; Lorenzo Stella; Margherita Silengo; Bruno Dallapiccola; Marco Tartaglia

Myhre syndrome is a developmental disorder characterized by reduced growth, generalized muscular hypertrophy, facial dysmorphism, deafness, cognitive deficits, joint stiffness, and skeletal anomalies. Here, by performing exome sequencing of a single affected individual and coupling the results to a hypothesis-driven filtering strategy, we establish that heterozygous mutations in SMAD4, which encodes for a transducer mediating transforming growth factor β and bone morphogenetic protein signaling branches, underlie this rare Mendelian trait. Two recurrent de novo SMAD4 mutations were identified in eight unrelated subjects. Both mutations were missense changes altering Ile500 within the evolutionary conserved MAD homology 2 domain, a well known mutational hot spot in malignancies. Structural analyses suggest that the substituted residues are likely to perturb the binding properties of the mutant protein to signaling partners. Although SMAD4 has been established as a tumor suppressor gene somatically mutated in pancreatic, gastrointestinal, and skin cancers, and germline loss-of-function lesions and deletions of this gene have been documented to cause disorders that predispose individuals to gastrointestinal cancer and vascular dysplasias, the present report identifies a previously unrecognized class of mutations in the gene with profound impact on development and growth.


American Journal of Human Genetics | 2015

Mutations Impairing GSK3-Mediated MAF Phosphorylation Cause Cataract, Deafness, Intellectual Disability, Seizures, and a Down Syndrome-like Facies

Marcello Niceta; Emilia Stellacci; Karen W. Gripp; Giuseppe Zampino; Maria Kousi; Massimiliano Anselmi; Alice Traversa; Andrea Ciolfi; Deborah L. Stabley; Alessandro Bruselles; Viviana Caputo; Serena Cecchetti; Sabrina Prudente; Maria Teresa Fiorenza; Carla Boitani; Nicole Philip; Dmitriy Niyazov; Chiara Leoni; Takaya Nakane; Kim M. Keppler-Noreuil; Stephen R. Braddock; Gabriele Gillessen-Kaesbach; Antonio Palleschi; Philippe M. Campeau; Brendan Lee; Celio Pouponnot; Lorenzo Stella; Gianfranco Bocchinfuso; Nicholas Katsanis; Katia Sol-Church

Transcription factors operate in developmental processes to mediate inductive events and cell competence, and perturbation of their function or regulation can dramatically affect morphogenesis, organogenesis, and growth. We report that a narrow spectrum of amino-acid substitutions within the transactivation domain of the v-maf avian musculoaponeurotic fibrosarcoma oncogene homolog (MAF), a leucine zipper-containing transcription factor of the AP1 superfamily, profoundly affect development. Seven different de novo missense mutations involving conserved residues of the four GSK3 phosphorylation motifs were identified in eight unrelated individuals. The distinctive clinical phenotype, for which we propose the eponym Aymé-Gripp syndrome, is not limited to lens and eye defects as previously reported for MAF/Maf loss of function but includes sensorineural deafness, intellectual disability, seizures, brachycephaly, distinctive flat facial appearance, skeletal anomalies, mammary gland hypoplasia, and reduced growth. Disease-causing mutations were demonstrated to impair proper MAF phosphorylation, ubiquitination and proteasomal degradation, perturbed gene expression in primary skin fibroblasts, and induced neurodevelopmental defects in an in vivo model. Our findings nosologically and clinically delineate a previously poorly understood recognizable multisystem disorder, provide evidence for MAF governing a wider range of developmental programs than previously appreciated, and describe a novel instance of protein dosage effect severely perturbing development.


American Journal of Human Genetics | 2016

Biallelic Mutations in TBCD, Encoding the Tubulin Folding Cofactor D, Perturb Microtubule Dynamics and Cause Early-Onset Encephalopathy

Elisabetta Flex; Marcello Niceta; Serena Cecchetti; Isabelle Thiffault; Margaret G. Au; Alessandro Capuano; Emanuela Piermarini; Anna A. Ivanova; Joshua W. Francis; Giovanni Chillemi; Balasubramanian Chandramouli; Giovanna Carpentieri; Charlotte A. Haaxma; Andrea Ciolfi; Simone Pizzi; Ganka Douglas; Kara L. Levine; Antonella Sferra; Maria Lisa Dentici; Rolph Pfundt; Jean Baptiste Le Pichon; Emily Farrow; Frank Baas; Fiorella Piemonte; Bruno Dallapiccola; John M. Graham; Carol J. Saunders; Enrico Bertini; Richard A. Kahn; David A. Koolen

Microtubules are dynamic cytoskeletal elements coordinating and supporting a variety of neuronal processes, including cell division, migration, polarity, intracellular trafficking, and signal transduction. Mutations in genes encoding tubulins and microtubule-associated proteins are known to cause neurodevelopmental and neurodegenerative disorders. Growing evidence suggests that altered microtubule dynamics may also underlie or contribute to neurodevelopmental disorders and neurodegeneration. We report that biallelic mutations in TBCD, encoding one of the five co-chaperones required for assembly and disassembly of the αβ-tubulin heterodimer, the structural unit of microtubules, cause a disease with neurodevelopmental and neurodegenerative features characterized by early-onset cortical atrophy, secondary hypomyelination, microcephaly, thin corpus callosum, developmental delay, intellectual disability, seizures, optic atrophy, and spastic quadriplegia. Molecular dynamics simulations predicted long-range and/or local structural perturbations associated with the disease-causing mutations. Biochemical analyses documented variably reduced levels of TBCD, indicating relative instability of mutant proteins, and defective β-tubulin binding in a subset of the tested mutants. Reduced or defective TBCD function resulted in decreased soluble α/β-tubulin levels and accelerated microtubule polymerization in fibroblasts from affected subjects, demonstrating an overall shift toward a more rapidly growing and stable microtubule population. These cells displayed an aberrant mitotic spindle with disorganized, tangle-shaped microtubules and reduced aster formation, which however did not alter appreciably the rate of cell proliferation. Our findings establish that defective TBCD function underlies a recognizable encephalopathy and drives accelerated microtubule polymerization and enhanced microtubule stability, underscoring an additional cause of altered microtubule dynamics with impact on neuronal function and survival in the developing brain.


Journal of Neurology | 2014

Childhood onset tubular aggregate myopathy associated with de novo STIM1 mutations

Carola Hedberg; Marcello Niceta; Fabiana Fattori; Björn Lindvall; Andrea Ciolfi; Adele D'Amico; Giorgio A. Tasca; Stefania Petrini; Mar Tulinius; Marco Tartaglia; Anders Oldfors; Enrico Bertini

Abstract We investigated three unrelated patients with tubular-aggregate myopathy and slowly progressive muscle weakness manifesting in the first years of life. All patients showed type 1 muscle fiber predominance and hypotrophy of type 2 fibers. Tubular aggregates were abundant. In all three patients mutations were identified in the gene STIM1, and the mutations were found to be de novo in all patients. In one of the patients the mutation was identified by exome sequencing. Two patients harbored the previously described mutation c.326A>G p.(His109Arg), while the third patient had a novel mutation c.343A>T p.(Ile115Phe). Taking our series together with previously published cases, the c.326A>G p.(His109Arg) seems to be a hotspot mutation that is characteristically related to early onset muscle weakness.


American Journal of Human Genetics | 2016

TBCE Mutations Cause Early-Onset Progressive Encephalopathy with Distal Spinal Muscular Atrophy

Antonella Sferra; Gilbert Baillat; Teresa Rizza; Sabina Barresi; Elisabetta Flex; Giorgio A. Tasca; Adele D’Amico; Emanuele Bellacchio; Andrea Ciolfi; Viviana Caputo; Serena Cecchetti; Annalaura Torella; Ginevra Zanni; Daria Diodato; Emanuela Piermarini; Marcello Niceta; Antonietta Coppola; Enrico Tedeschi; Diego Martinelli; Carlo Dionisi-Vici; Vincenzo Nigro; Bruno Dallapiccola; Claudia Compagnucci; Marco Tartaglia; Georg Haase; Enrico Bertini

Tubulinopathies constitute a family of neurodevelopmental/neurodegenerative disorders caused by mutations in several genes encoding tubulin isoforms. Loss-of-function mutations in TBCE, encoding one of the five tubulin-specific chaperones involved in tubulin folding and polymerization, cause two rare neurodevelopmental syndromes, hypoparathyroidism-retardation-dysmorphism and Kenny-Caffey syndrome. Although a missense mutation in Tbce has been associated with progressive distal motor neuronopathy in the pmn/pmn mice, no similar degenerative phenotype has been recognized in humans. We report on the identification of an early-onset and progressive neurodegenerative encephalopathy with distal spinal muscular atrophy resembling the phenotype of pmn/pmn mice and caused by biallelic TBCE mutations, with the c.464T>A (p.Ile155Asn) change occurring at the heterozygous/homozygous state in six affected subjects from four unrelated families originated from the same geographical area in Southern Italy. Western blot analysis of patient fibroblasts documented a reduced amount of TBCE, suggestive of rapid degradation of the mutant protein, similarly to what was observed in pmn/pmn fibroblasts. The impact of TBCE mutations on microtubule polymerization was determined using biochemical fractionation and analyzing the nucleation and growth of microtubules at the centrosome and extracentrosomal sites after treatment with nocodazole. Primary fibroblasts obtained from affected subjects displayed a reduced level of polymerized α-tubulin, similarly to tail fibroblasts of pmn/pmn mice. Moreover, markedly delayed microtubule re-polymerization and abnormal mitotic spindles with disorganized microtubule arrangement were also documented. Although loss of function of TBCE has been documented to impact multiple developmental processes, the present findings provide evidence that hypomorphic TBCE mutations primarily drive neurodegeneration.


Journal of Neurology | 2017

Novel mutations in IBA57 are associated with leukodystrophy and variable clinical phenotypes

Alessandra Torraco; Anna Ardissone; Federica Invernizzi; Teresa Rizza; Giuseppe Fiermonte; Marcello Niceta; Nadia Zanetti; Diego Martinelli; Angelo Vozza; Daniela Verrigni; Michela Di Nottia; Eleonora Lamantea; Daria Diodato; Marco Tartaglia; Carlo Dionisi-Vici; Isabella Moroni; Laura Farina; Enrico Bertini; Daniele Ghezzi; Rosalba Carrozzo

Defects of the Fe/S cluster biosynthesis represent a subgroup of diseases affecting the mitochondrial energy metabolism. In the last years, mutations in four genes (NFU1, BOLA3, ISCA2 and IBA57) have been related to a new group of multiple mitochondrial dysfunction syndromes characterized by lactic acidosis, hyperglycinemia, multiple defects of the respiratory chain complexes, and impairment of four lipoic acid-dependent enzymes: α-ketoglutarate dehydrogenase complex, pyruvic dehydrogenase, branched-chain α-keto acid dehydrogenase complex and the H protein of the glycine cleavage system. Few patients have been reported with mutations in IBA57 and with variable clinical phenotype. Herein, we describe four unrelated patients carrying novel mutations in IBA57. All patients presented with combined or isolated defect of complex I and II. Clinical features varied widely, ranging from fatal infantile onset of the disease to acute and severe psychomotor regression after the first year of life. Brain MRI was characterized by cavitating leukodystrophy. The identified mutations were never reported previously and all had a dramatic effect on IBA57 stability. Our study contributes to expand the array of the genotypic variation of IBA57 and delineates the leukodystrophic pattern of IBA57 deficient patients.


Clinical Genetics | 2017

Mutations in the IRBIT domain of ITPR1 are a frequent cause of autosomal dominant nonprogressive congenital ataxia.

Sabina Barresi; Marcello Niceta; Paolo Alfieri; V. Brankovic; Giorgia Piccini; Alessandro Bruselles; M.R. Barone; R. Cusmai; Marco Tartaglia; Enrico Bertini; Ginevra Zanni

Congenital ataxias are nonprogressive neurological disorders characterized by neonatal hypotonia, developmental delay and ataxia, variably associated with intellectual disability and other neurological or extraneurological features. We performed trio‐based whole‐exome sequencing of 12 families with congenital cerebellar and/or vermis atrophy in parallel with targeted next‐generation sequencing of known ataxia genes (CACNA1A, ITPR1, KCNC3, ATP2B3 and GRM1) in 12 additional patients with a similar phenotype. Novel pathological mutations of ITPR1 (inositol 1,4,5‐trisphosphate receptor, type 1) were found in seven patients from four families (4/24, ∼16.8%) all localized in the IRBIT (inositol triphosphate receptor binding protein) domain which plays an essential role in the regulation of neuronal plasticity and development. Our study expands the mutational spectrum of ITPR1‐related congenital ataxia and indicates that ITPR1 gene screening should be implemented in this subgroup of ataxias.


Clinical Genetics | 2017

A novel mutation in NDUFB11 unveils a new clinical phenotype associated with lactic acidosis and sideroblastic anemia

Alessandra Torraco; Marzia Bianchi; Daniela Verrigni; Vania Gelmetti; Lisa G. Riley; Marcello Niceta; Diego Martinelli; Arianna Montanari; Y. Guo; Teresa Rizza; Daria Diodato; M. Di Nottia; B. Lucarelli; F. Sorrentino; Fiorella Piemonte; Silvia Francisci; Marco Tartaglia; Enza Maria Valente; Carlo Dionisi-Vici; John Christodoulou; Enrico Bertini; Rosalba Carrozzo

NDUFB11, a component of mitochondrial complex I, is a relatively small integral membrane protein, belonging to the “supernumerary” group of subunits, but proved to be absolutely essential for the assembly of an active complex I. Mutations in the X‐linked nuclear‐encoded NDUFB11 gene have recently been discovered in association with two distinct phenotypes, i.e. microphthalmia with linear skin defects and histiocytoid cardiomyopathy. We report on a male with complex I deficiency, caused by a de novo mutation in NDUFB11 and displaying early‐onset sideroblastic anemia as the unique feature. This is the third report that describes a mutation in NDUFB11, but all are associated with a different phenotype. Our results further expand the molecular spectrum and associated clinical phenotype of NDUFB11 defects.


Clinical Genetics | 2017

Microcephaly, intractable seizures and developmental delay caused by biallelic variants in TBCD: Further delineation of a new chaperone-mediated tubulinopathy

Ben Pode-Shakked; Hila Barash; Limor Ziv; Karen W. Gripp; Elisabetta Flex; Ortal Barel; Karen S. Carvalho; Mena Scavina; Giovanni Chillemi; Marcello Niceta; Eran Eyal; Nitzan Kol; Bruria Ben-Zeev; O. Bar-Yosef; Dina Marek-Yagel; Enrico Bertini; Angela L. Duker; Yair Anikster; Marco Tartaglia; Annick Raas-Rothschild

Microtubule dynamics play a crucial role in neuronal development and function, and several neurodevelopmental disorders have been linked to mutations in genes encoding tubulins and functionally related proteins. Most recently, variants in the tubulin cofactor D (TBCD) gene, which encodes one of the five co‐chaperones required for assembly and disassembly of α/β‐tubulin heterodimer, were reported to underlie a recessive neurodevelopmental/neurodegenerative disorder. We report on five patients from three unrelated families, who presented with microcephaly, intellectual disability, intractable seizures, optic nerve pallor/atrophy, and cortical atrophy with delayed myelination and thinned corpus callosum on brain imaging. Exome sequencing allowed the identification of biallelic variants in TBCD segregating with the disease in the three families. TBCD protein level was significantly reduced in cultured fibroblasts from one patient, supporting defective TBCD function as the event underlying the disorder. Such reduced expression was associated with accelerated microtubule re‐polymerization. Morpholino‐mediated TBCD knockdown in zebrafish recapitulated several key pathological features of the human disease, and TBCD overexpression in the same model confirmed previous studies documenting an obligate dependency on proper TBCD levels during development. Our findings confirm the link between inactivating TBCD variants and this newly described chaperone‐associated tubulinopathy, and provide insights into the phenotype of this disorder.


Journal of Inherited Metabolic Disease | 2016

Expanding the molecular diversity and phenotypic spectrum of glycerol 3-phosphate dehydrogenase 1 deficiency

Carlo Dionisi-Vici; Eyal Shteyer; Marcello Niceta; Cristiano Rizzo; Ben Pode-Shakked; Giovanni Chillemi; Alessandro Bruselles; Michela Semeraro; Ortal Barel; Eran Eyal; Nitzan Kol; Yael Haberman; Avishai Lahad; Francesca Diomedi-Camassei; Dina Marek-Yagel; Gideon Rechavi; Marco Tartaglia; Yair Anikster

Transient infantile hypertriglyceridemia (HTGT1; OMIM #614480) is a rare autosomal recessive disorder, which manifests in early infancy with transient hypertriglyceridemia, hepatomegaly, elevated liver enzymes, persistent fatty liver and hepatic fibrosis. This rare clinical entity is caused by inactivating mutations in the GPD1 gene, which encodes the cytosolic isoform of glycerol-3-phosphate dehydrogenase. Here we report on four patients from three unrelated families of diverse ethnic origins, who presented with hepatomegaly, liver steatosis, hypertriglyceridemia, with or without fasting ketotic hypoglycemia. Whole exome sequencing revealed the affected individuals to harbor deleterious biallelic mutations in the GPD1 gene, including the previously undescribed c.806G > A (p.Arg269Gln) and c.640T > C (p.Cys214Arg) mutations. The clinical features in three of our patients showed several differences compared to the original reports. One subject presented with recurrent episodes of fasting hypoglycemia along with hepatomegaly, hypetriglyceridemia, and elevated liver enzymes; the second showed a severe liver disease, with intrahepatic cholestasis associated with kidney involvement; finally, the third presented persistent hypertriglyceridemia at the age of 30 years. These findings expand the current knowledge of this rare disorder, both with regard to the phenotype and molecular basis. The enlarged phenotypic spectrum of glycerol-3-phosphate dehydrogenase 1 deficiency can mimic other inborn errors of metabolism with liver involvement and should alert clinicians to recognize this entity by considering GPD1 mutations in appropriate clinical settings.

Collaboration


Dive into the Marcello Niceta's collaboration.

Top Co-Authors

Avatar

Marco Tartaglia

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Enrico Bertini

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Andrea Ciolfi

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar

Rosalba Carrozzo

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Teresa Rizza

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daria Diodato

Boston Children's Hospital

View shared research outputs
Top Co-Authors

Avatar

Alessandro Bruselles

Istituto Superiore di Sanità

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniela Verrigni

Boston Children's Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge