Network


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

Hotspot


Dive into the research topics where Niccolo E. Mencacci is active.

Publication


Featured researches published by Niccolo E. Mencacci.


JAMA Neurology | 2014

Long-term Clinical Outcome of Fetal Cell Transplantation for Parkinson Disease: Two Case Reports

Zinovia Kefalopoulou; Marios Politis; Paola Piccini; Niccolo E. Mencacci; Kailash P. Bhatia; Marjan Jahanshahi; Håkan Widner; Stig Rehncrona; Patrik Brundin; Anders Björklund; Olle Lindvall; Patricia Limousin; Niall Quinn; Thomas Foltynie

IMPORTANCE Recent advances in stem cell technologies have rekindled an interest in the use of cell replacement strategies for patients with Parkinson disease. This study reports the very long-term clinical outcomes of fetal cell transplantation in 2 patients with Parkinson disease. Such long-term follow-up data can usefully inform on the potential efficacy of this approach, as well as the design of trials for its further evaluation. OBSERVATIONS Two patients received intrastriatal grafts of human fetal ventral mesencephalic tissue, rich in dopaminergic neuroblasts, as restorative treatment for their Parkinson disease. To evaluate the very long-term efficacy of the grafts, clinical assessments were performed 18 and 15 years posttransplantation. Motor improvements gained gradually over the first postoperative years were sustained up to 18 years posttransplantation, while both patients have discontinued, and remained free of any, pharmacological dopaminergic therapy. CONCLUSIONS AND RELEVANCE The results from these 2 cases indicate that dopaminergic cell transplantation can offer very long-term symptomatic relief in patients with Parkinson disease and provide proof-of-concept support for future clinical trials using fetal or stem cell therapies.


Brain | 2013

Glucocerebrosidase mutations influence the natural history of Parkinson's disease in a community-based incident cohort

Sophie Winder-Rhodes; Jonathan R. Evans; Maria Ban; Sarah Mason; Caroline H. Williams-Gray; Thomas Foltynie; Raquel Duran; Niccolo E. Mencacci; Stephen Sawcer; Roger A. Barker

Carriers of mutations in the glucocerebrosidase gene (GBA) are at increased risk of developing Parkinsons disease. The frequency of GBA mutations in unselected Parkinsons disease populations has not been established. Furthermore, no previous studies have investigated the influence of GBA mutations on the natural history of Parkinsons disease using prospective follow-up. We studied DNA from 262 cases who had been recruited at diagnosis into one of two independent community-based incidence studies of Parkinsons disease. In 121 cases, longitudinal data regarding progression of motor disability and cognitive function were derived from follow-up assessments conducted every 18 months for a median of 71 months. Sequencing of the GBA was performed after two-stage polymerase chain reaction amplification. The carrier frequency of genetic variants in GBA was determined. Baseline demographic and clinical variables were compared between cases who were either GBA mutation carriers, polymorphism carriers or wild-type homozygotes. Cox regression analysis was used to model progression to major motor (Hoehn and Yahr stage 3), and cognitive (dementia) end-points in cases followed longitudinally. We show that in a representative, unselected UK Parkinsons disease population, GBA mutations are present at a frequency of 3.5%. This is higher than the prevalence of other genetic mutations currently associated with Parkinsons disease and indicates that GBA mutations make an important contribution to Parkinsons disease encountered in the community setting. Baseline clinical characteristics did not differ significantly between cases with and without GBA sequence variants. However, the hazard ratio for progression both to dementia (5.7, P = 0.003) and Hoehn and Yahr stage 3 (4.2, P = 0.003) were significantly greater in GBA mutation carriers. We also show that carriers of polymorphisms in GBA which are not generally considered to increase Parkinsons disease risk are at significantly increased risk of progression to Hoehn and Yahr stage 3 (3.2, P = 0.004). Our results indicate that genetic variation in GBA has an important impact on the natural history of Parkinsons disease. To our knowledge, this is the first time a genetic locus has been shown to influence motor progression in Parkinsons disease. If confirmed in further studies, this may indicate that GBA mutation status could be used as a prognostic marker in Parkinsons disease. Elucidation of the molecular mechanisms that underlie this effect will further our understanding of the pathogenesis of the disease and may in turn suggest novel therapeutic strategies.


Annals of Neurology | 2013

Mutations in the autoregulatory domain of β-tubulin 4a cause hereditary dystonia

Joshua Hersheson; Niccolo E. Mencacci; Mary B. Davis; Nicola MacDonald; Daniah Trabzuni; Mina Ryten; Alan Pittman; Reema Paudel; Eleanna Kara; Katherine Fawcett; Vincent Plagnol; Kailash P. Bhatia; Alan Medlar; Horia Stanescu; John Hardy; Robert Kleta; Nicholas W. Wood; Henry Houlden

Dystonia type 4 (DYT4) was first described in a large family from Heacham in Norfolk with an autosomal dominantly inherited whispering dysphonia, generalized dystonia, and a characteristic hobby horse ataxic gait. We carried out a genetic linkage analysis in the extended DYT4 family that spanned 7 generations from England and Australia, revealing a single LOD score peak of 6.33 on chromosome 19p13.12‐13. Exome sequencing in 2 cousins identified a single cosegregating mutation (p.R2G) in the β‐tubulin 4a (TUBB4a) gene that was absent in a large number of controls. The mutation is highly conserved in the β‐tubulin autoregulatory MREI (methionine–arginine–glutamic acid–isoleucine) domain, highly expressed in the central nervous system, and extensive in vitro work has previously demonstrated that substitutions at residue 2, specifically R2G, disrupt the autoregulatory capability of the wild‐type β‐tubulin peptide, affirming the role of the cytoskeleton in dystonia pathogenesis. Ann Neurol 2013;73:546–553


Brain | 2014

Parkinson's disease in GTP cyclohydrolase 1 mutation carriers

Niccolo E. Mencacci; Ioannis U. Isaias; Martin M. Reich; Christos Ganos; Vincent Plagnol; James M. Polke; Jose Bras; Joshua Hersheson; Maria Stamelou; Alan Pittman; Alastair J. Noyce; Kin Mok; Thomas Opladen; Erdmute Kunstmann; Sybille Hodecker; Alexander Münchau; Jens Volkmann; Samuel Samnick; Katie Sidle; Tina Nanji; Mary G. Sweeney; Henry Houlden; Amit Batla; Anna Zecchinelli; Gianni Pezzoli; Giorgio Marotta; Andrew J. Lees; Paulo Alegria; Paul Krack; Florence Cormier-Dequaire

Mutations in the gene encoding the dopamine-synthetic enzyme GTP cyclohydrolase-1 (GCH1) cause DOPA-responsive dystonia (DRD). Mencacci et al. demonstrate that GCH1 variants are associated with an increased risk of Parkinsons disease in both DRD pedigrees and in patients with Parkinsons disease but without a family history of DRD.


JAMA Neurology | 2013

Parkin disease: A clinicopathologic entity?

Karen M. Doherty; Laura Silveira-Moriyama; Laura Parkkinen; Daniel G. Healy; Michael Farrell; Niccolo E. Mencacci; Zeshan Ahmed; Francesca Brett; John Hardy; Niall Quinn; Timothy J. Counihan; Timothy Lynch; Zoe Fox; Tamas Revesz; Andrew J. Lees; Janice L. Holton

IMPORTANCE Mutations in the gene encoding parkin (PARK2) are the most common cause of autosomal recessive juvenile-onset and young-onset parkinsonism. The few available detailed neuropathologic reports suggest that homozygous and compound heterozygous parkin mutations are characterized by severe substantia nigra pars compacta neuronal loss. OBJECTIVE To investigate whether parkin-linked parkinsonism is a different clinicopathologic entity to Parkinson disease (PD). DESIGN, SETTING, AND PARTICIPANTS We describe the clinical, genetic, and neuropathologic findings of 5 unrelated cases of parkin disease and compare them with 5 pathologically confirmed PD cases and 4 control subjects. The PD control cases and normal control subjects were matched first for age at death then disease duration (PD only) for comparison. RESULTS Presenting signs in the parkin disease cases were hand or leg tremor often combined with dystonia. Mean age at onset was 34 years; all cases were compound heterozygous for mutations of parkin. Freezing of gait, postural deformity, and motor fluctuations were common late features. No patients had any evidence of cognitive impairment or dementia. Neuronal counts in the substantia nigra pars compacta revealed that neuronal loss in the parkin cases was as severe as that seen in PD, but relative preservation of the dorsal tier was seen in comparison with PD (P = .04). Mild neuronal loss was identified in the locus coeruleus and dorsal motor nucleus of the vagus, but not in the nucleus basalis of Meynert, raphe nucleus, or other brain regions. Sparse Lewy bodies were identified in 2 cases (brainstem and cortex). CONCLUSIONS AND RELEVANCE These findings support the notion that parkin disease is characterized by a more restricted morphologic abnormality than is found in PD, with predominantly ventral nigral degeneration and absent or rare Lewy bodies.


Movement Disorders | 2013

The Glucocerobrosidase E326K Variant Predisposes to Parkinson’s Disease, But Does Not Cause Gaucher’s Disease

Raquel Duran; Niccolo E. Mencacci; A Angeli; Maryam Shoai; Emma Deas; Henry Houlden; Atul Mehta; Derralynn Hughes; Timothy M. Cox; Patrick Deegan; A. H. V. Schapira; Andrew J. Lees; Patricia Limousin; Paul R. Jarman; Kailash P. Bhatia; Nicholas W. Wood; John Hardy; Thomas Foltynie

Heterozygous loss‐of‐function mutations in the acid beta‐glucocerebrosidase (GBA1) gene, responsible for the recessive lysosomal storage disorder, Gauchers disease (GD), are the strongest known risk factor for Parkinsons disease (PD). Our aim was to assess the contribution of GBA1 mutations in a series of early‐onset PD.


Cell Death and Disease | 2016

Mitofusin-mediated ER stress triggers neurodegeneration in pink1/parkin models of Parkinson's disease.

I Celardo; A C Costa; Susann Lehmann; Carolyn Jones; Nicholas W. Wood; Niccolo E. Mencacci; Giovanna R. Mallucci; Shy Loh; L M Martins

Mutations in PINK1 and PARKIN cause early-onset Parkinson’s disease (PD), thought to be due to mitochondrial toxicity. Here, we show that in Drosophila pink1 and parkin mutants, defective mitochondria also give rise to endoplasmic reticulum (ER) stress signalling, specifically to the activation of the protein kinase R-like endoplasmic reticulum kinase (PERK) branch of the unfolded protein response (UPR). We show that enhanced ER stress signalling in pink1 and parkin mutants is mediated by mitofusin bridges, which occur between defective mitochondria and the ER. Reducing mitofusin contacts with the ER is neuroprotective, through suppression of PERK signalling, while mitochondrial dysfunction remains unchanged. Further, both genetic inhibition of dPerk-dependent ER stress signalling and pharmacological inhibition using the PERK inhibitor GSK2606414 were neuroprotective in both pink1 and parkin mutants. We conclude that activation of ER stress by defective mitochondria is neurotoxic in pink1 and parkin flies and that the reduction of this signalling is neuroprotective, independently of defective mitochondria. A video abstract for this article is available online in the supplementary information


Neurology | 2016

A genome-wide association study in multiple system atrophy

Anna Sailer; Sonja W. Scholz; Michael A. Nalls; Claudia Schulte; Monica Federoff; T. Ryan Price; Andrew J. Lees; Owen A. Ross; Dennis W. Dickson; Kin Mok; Niccolo E. Mencacci; Lucia Schottlaender; Viorica Chelban; Helen Ling; Sean S. O'Sullivan; Nicholas W. Wood; Bryan J. Traynor; Luigi Ferrucci; Howard J. Federoff; Timothy R. Mhyre; Huw R. Morris; Günther Deuschl; Niall Quinn; Håkan Widner; Alberto Albanese; Jon Infante; Kailash P. Bhatia; Werner Poewe; Wolfgang H. Oertel; Günter U. Höglinger

Objective: To identify genetic variants that play a role in the pathogenesis of multiple system atrophy (MSA), we undertook a genome-wide association study (GWAS). Methods: We performed a GWAS with >5 million genotyped and imputed single nucleotide polymorphisms (SNPs) in 918 patients with MSA of European ancestry and 3,864 controls. MSA cases were collected from North American and European centers, one third of which were neuropathologically confirmed. Results: We found no significant loci after stringent multiple testing correction. A number of regions emerged as potentially interesting for follow-up at p < 1 × 10−6, including SNPs in the genes FBXO47, ELOVL7, EDN1, and MAPT. Contrary to previous reports, we found no association of the genes SNCA and COQ2 with MSA. Conclusions: We present a GWAS in MSA. We have identified several potentially interesting gene loci, including the MAPT locus, whose significance will have to be evaluated in a larger sample set. Common genetic variation in SNCA and COQ2 does not seem to be associated with MSA. In the future, additional samples of well-characterized patients with MSA will need to be collected to perform a larger MSA GWAS, but this initial study forms the basis for these next steps.


Neurology | 2015

ADCY5 mutations are another cause of benign hereditary chorea

Niccolo E. Mencacci; Roberto Erro; Sarah Wiethoff; Joshua Hersheson; Mina Ryten; Bettina Balint; Christos Ganos; Maria Stamelou; Niall Quinn; Henry Houlden; Nicholas W. Wood; Kailash P. Bhatia

Objective: To determine the contribution of ADCY5 mutations in cases with genetically undefined benign hereditary chorea (BHC). Methods: We studied 18 unrelated cases with BHC (7 familial, 11 sporadic) who were negative for NKX2-1 mutations. The diagnosis of BHC was based on the presence of a childhood-onset movement disorder, predominantly characterized by chorea and no other major neurologic features. ADCY5 analysis was performed by whole-exome sequencing or Sanger sequencing. ADCY5 and NKX2-1 expression during brain development and in the adult human brain was assessed using microarray analysis of postmortem brain tissue. Results: The c.1252C>T; p.R418W mutation was identified in 2 cases (1 familial, 1 sporadic). The familial case inherited the mutation from the affected father, who had a much milder presentation, likely due to low-grade somatic mosaicism. The mutation was de novo in the sporadic case. The clinical presentation of these cases featured nonparoxysmal generalized chorea, as well as dystonia in the most severely affected, but no facial myokymia. We observed significant progression of symptoms in ADCY5 mutation carriers, in contrast to BHC secondary to NKX2-1 mutations. The difference in the clinical course is mirrored by the brain expression data, showing increasing ADCY5 expression in the striatum during brain development, whereas NKX2-1 shows an opposite trend. Conclusions: Our study identifies mutations in ADCY5, the gene previously linked to familial dyskinesia with facial myokymia, as a cause of familial and sporadic BHC. ADCY5 genetic analysis should be performed in cases with a benign choreiform movement disorder even in the absence of facial myokymia.


American Journal of Human Genetics | 2015

A Missense Mutation in KCTD17 Causes Autosomal Dominant Myoclonus-Dystonia

Niccolo E. Mencacci; Ignacio Rubio-Agusti; Anselm A. Zdebik; Friedrich Asmus; Marthe H.R. Ludtmann; Mina Ryten; Vincent Plagnol; Ann-Kathrin Hauser; Sara Bandres-Ciga; Conceição Bettencourt; Paola Forabosco; Deborah Hughes; Marc M.P. Soutar; Kathryn J. Peall; Huw R. Morris; Daniah Trabzuni; Mehmet Tekman; Horia Stanescu; Robert Kleta; Miryam Carecchio; Giovanna Zorzi; Nardo Nardocci; Barbara Garavaglia; Ebba Lohmann; Anne Weissbach; Christine Klein; John Hardy; Alan Pittman; Thomas Foltynie; Andrey Y. Abramov

Myoclonus-dystonia (M-D) is a rare movement disorder characterized by a combination of non-epileptic myoclonic jerks and dystonia. SGCE mutations represent a major cause for familial M-D being responsible for 30%-50% of cases. After excluding SGCE mutations, we identified through a combination of linkage analysis and whole-exome sequencing KCTD17 c.434 G>A p.(Arg145His) as the only segregating variant in a dominant British pedigree with seven subjects affected by M-D. A subsequent screening in a cohort of M-D cases without mutations in SGCE revealed the same KCTD17 variant in a German family. The clinical presentation of the KCTD17-mutated cases was distinct from the phenotype usually observed in M-D due to SGCE mutations. All cases initially presented with mild myoclonus affecting the upper limbs. Dystonia showed a progressive course, with increasing severity of symptoms and spreading from the cranio-cervical region to other sites. KCTD17 is abundantly expressed in all brain regions with the highest expression in the putamen. Weighted gene co-expression network analysis, based on mRNA expression profile of brain samples from neuropathologically healthy individuals, showed that KCTD17 is part of a putamen gene network, which is significantly enriched for dystonia genes. Functional annotation of the network showed an over-representation of genes involved in post-synaptic dopaminergic transmission. Functional studies in mutation bearing fibroblasts demonstrated abnormalities in endoplasmic reticulum-dependent calcium signaling. In conclusion, we demonstrate that the KCTD17 c.434 G>A p.(Arg145His) mutation causes autosomal dominant M-D. Further functional studies are warranted to further characterize the nature of KCTD17 contribution to the molecular pathogenesis of M-D.

Collaboration


Dive into the Niccolo E. Mencacci's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicholas W. Wood

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Henry Houlden

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

John Hardy

University College London

View shared research outputs
Top Co-Authors

Avatar

Andrew J. Lees

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar

Amit Batla

University College London

View shared research outputs
Top Co-Authors

Avatar

Alan Pittman

University College London

View shared research outputs
Top Co-Authors

Avatar

Thomas Foltynie

UCL Institute of Neurology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Maria Stamelou

National and Kapodistrian University of Athens

View shared research outputs
Researchain Logo
Decentralizing Knowledge