Network


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

Hotspot


Dive into the research topics where Paola Ghiorzo is active.

Publication


Featured researches published by Paola Ghiorzo.


Nature Genetics | 2009

Genome-wide association study identifies three loci associated with melanoma risk

D. Timothy Bishop; Florence Demenais; Mark M. Iles; Mark Harland; John C. Taylor; Eve Corda; Juliette Randerson-Moor; Joanne F. Aitken; Marie-Françoise Avril; Esther Azizi; Bert Bakker; Giovanna Bianchi-Scarrà; Brigitte Bressac-de Paillerets; Donato Calista; Lisa A. Cannon-Albright; Thomas Chin-a-Woeng; Tadeusz Dębniak; Gilli Galore-Haskel; Paola Ghiorzo; Ivo Gut; Johan Hansson; Marko Hocevar; Veronica Höiom; John L. Hopper; Christian Ingvar; Peter A. Kanetsky; Richard Kefford; Maria Teresa Landi; Julie Lang; Jan Lubinski

We report a genome-wide association study of melanoma conducted by the GenoMEL consortium based on 317K tagging SNPs for 1,650 selected cases and 4,336 controls, with replication in an additional two cohorts (1,149 selected cases and 964 controls from GenoMEL, and a population-based case-control study in Leeds of 1,163 cases and 903 controls). The genome-wide screen identified five loci with genotyped or imputed SNPs reaching P < 5 × 10−7. Three of these loci were replicated: 16q24 encompassing MC1R (combined P = 2.54 × 10−27 for rs258322), 11q14-q21 encompassing TYR (P = 2.41 × 10−14 for rs1393350) and 9p21 adjacent to MTAP and flanking CDKN2A (P = 4.03 × 10−7 for rs7023329). MC1R and TYR are associated with pigmentation, freckling and cutaneous sun sensitivity, well-recognized melanoma risk factors. Common variants within the 9p21 locus have not previously been associated with melanoma. Despite wide variation in allele frequency, these genetic variants show notable homogeneity of effect across populations of European ancestry living at different latitudes and show independent association to disease risk.


Cancer Research | 2006

High-risk melanoma susceptibility genes and pancreatic cancer, neural system tumors, and uveal melanoma across GenoMEL

Alisa M. Goldstein; May Chan; Mark Harland; Elizabeth M. Gillanders; Nicholas K. Hayward; Marie-Françoise Avril; Esther Azizi; Giovanna Bianchi-Scarrà; D. Timothy Bishop; Brigitte Bressac-de Paillerets; William Bruno; Donato Calista; Lisa A. Cannon Albright; Florence Demenais; David E. Elder; Paola Ghiorzo; Nelleke A. Gruis; Johan Hansson; David Hogg; Elizabeth A. Holland; Peter A. Kanetsky; Richard F. Kefford; Maria Teresa Landi; Julie Lang; Sancy A. Leachman; Rona M. MacKie; Veronica Magnusson; Graham J. Mann; Kristin B. Niendorf; Julia A. Newton Bishop

GenoMEL, comprising major familial melanoma research groups from North America, Europe, Asia, and Australia has created the largest familial melanoma sample yet available to characterize mutations in the high-risk melanoma susceptibility genes CDKN2A/alternate reading frames (ARF), which encodes p16 and p14ARF, and CDK4 and to evaluate their relationship with pancreatic cancer (PC), neural system tumors (NST), and uveal melanoma (UM). This study included 466 families (2,137 patients) with at least three melanoma patients from 17 GenoMEL centers. Overall, 41% (n = 190) of families had mutations; most involved p16 (n = 178). Mutations in CDK4 (n = 5) and ARF (n = 7) occurred at similar frequencies (2-3%). There were striking differences in mutations across geographic locales. The proportion of families with the most frequent founder mutation(s) of each locale differed significantly across the seven regions (P = 0.0009). Single founder CDKN2A mutations were predominant in Sweden (p.R112_L113insR, 92% of familys mutations) and the Netherlands (c.225_243del19, 90% of familys mutations). France, Spain, and Italy had the same most frequent mutation (p.G101W). Similarly, Australia and United Kingdom had the same most common mutations (p.M53I, c.IVS2-105A>G, p.R24P, and p.L32P). As reported previously, there was a strong association between PC and CDKN2A mutations (P < 0.0001). This relationship differed by mutation. In contrast, there was little evidence for an association between CDKN2A mutations and NST (P = 0.52) or UM (P = 0.25). There was a marginally significant association between NST and ARF (P = 0.05). However, this particular evaluation had low power and requires confirmation. This GenoMEL study provides the most extensive characterization of mutations in high-risk melanoma susceptibility genes in families with three or more melanoma patients yet available.


Nature | 2011

A SUMOylation-defective MITF germline mutation predisposes to melanoma and renal carcinoma

Corine Bertolotto; Fabienne Lesueur; Sandy Giuliano; Thomas Strub; Mahaut de Lichy; Karine Bille; Philippe Dessen; Benoit d'Hayer; Hamida Mohamdi; Audrey Remenieras; Eve Maubec; Arnaud de la Fouchardière; Vincent Molinié; Pierre Vabres; Stéphane Dalle; Nicolas Poulalhon; Tanguy Martin-Denavit; Luc Thomas; Pascale Andry-Benzaquen; Nicolas Dupin; F. Boitier; Annick Rossi; Jean Luc Perrot; B. Labeille; Caroline Robert; Bernard Escudier; Olivier Caron; Laurence Brugières; Simon Saule; Betty Gardie

So far, no common environmental and/or phenotypic factor has been associated with melanoma and renal cell carcinoma (RCC). The known risk factors for melanoma include sun exposure, pigmentation and nevus phenotypes; risk factors associated with RCC include smoking, obesity and hypertension. A recent study of coexisting melanoma and RCC in the same patients supports a genetic predisposition underlying the association between these two cancers. The microphthalmia-associated transcription factor (MITF) has been proposed to act as a melanoma oncogene; it also stimulates the transcription of hypoxia inducible factor (HIF1A), the pathway of which is targeted by kidney cancer susceptibility genes. We therefore proposed that MITF might have a role in conferring a genetic predisposition to co-occurring melanoma and RCC. Here we identify a germline missense substitution in MITF (Mi-E318K) that occurred at a significantly higher frequency in genetically enriched patients affected with melanoma, RCC or both cancers, when compared with controls. Overall, Mi-E318K carriers had a higher than fivefold increased risk of developing melanoma, RCC or both cancers. Codon 318 is located in a small-ubiquitin-like modifier (SUMO) consensus site (ΨKXE) and Mi-E318K severely impaired SUMOylation of MITF. Mi-E318K enhanced MITF protein binding to the HIF1A promoter and increased its transcriptional activity compared to wild-type MITF. Further, we observed a global increase in Mi-E318K-occupied loci. In an RCC cell line, gene expression profiling identified a Mi-E318K signature related to cell growth, proliferation and inflammation. Lastly, the mutant protein enhanced melanocytic and renal cell clonogenicity, migration and invasion, consistent with a gain-of-function role in tumorigenesis. Our data provide insights into the link between SUMOylation, transcription and cancer.


Journal of Medical Genetics | 2006

Features associated with germline CDKN2A mutations: a GenoMEL study of melanoma-prone families from three continents

Alisa M. Goldstein; May Chan; Mark Harland; Nicholas K. Hayward; Florence Demenais; D. Timothy Bishop; Esther Azizi; Wilma Bergman; Giovanna Bianchi-Scarrà; William Bruno; Donato Calista; Lisa A. Cannon Albright; Valérie Chaudru; Agnès Chompret; Francisco Cuellar; David E. Elder; Paola Ghiorzo; Elizabeth M. Gillanders; Nelleke A. Gruis; Johan Hansson; David Hogg; Elizabeth A. Holland; Peter A. Kanetsky; Richard F. Kefford; Maria Teresa Landi; Julie Lang; Sancy A. Leachman; Rona M. MacKie; Veronica Magnusson; Graham J. Mann

Background: The major factors individually reported to be associated with an increased frequency of CDKN2A mutations are increased number of patients with melanoma in a family, early age at melanoma diagnosis, and family members with multiple primary melanomas (MPM) or pancreatic cancer. Methods: These four features were examined in 385 families with ⩾3 patients with melanoma pooled by 17 GenoMEL groups, and these attributes were compared across continents. Results: Overall, 39% of families had CDKN2A mutations ranging from 20% (32/162) in Australia to 45% (29/65) in North America to 57% (89/157) in Europe. All four features in each group, except pancreatic cancer in Australia (p = 0.38), individually showed significant associations with CDKN2A mutations, but the effects varied widely across continents. Multivariate examination also showed different predictors of mutation risk across continents. In Australian families, ⩾2 patients with MPM, median age at melanoma diagnosis ⩽40 years and ⩾6 patients with melanoma in a family jointly predicted the mutation risk. In European families, all four factors concurrently predicted the risk, but with less stringent criteria than in Australia. In North American families, only ⩾1 patient with MPM and age at diagnosis ⩽40 years simultaneously predicted the mutation risk. Conclusions: The variation in CDKN2A mutations for the four features across continents is consistent with the lower melanoma incidence rates in Europe and higher rates of sporadic melanoma in Australia. The lack of a pancreatic cancer–CDKN2A mutation relationship in Australia probably reflects the divergent spectrum of mutations in families from Australia versus those from North America and Europe. GenoMEL is exploring candidate host, genetic and/or environmental risk factors to better understand the variation observed.


Nature Genetics | 2008

Common sequence variants on 20q11.22 confer melanoma susceptibility

Kevin M. Brown; Stuart Macgregor; Grant W. Montgomery; David Craig; Zhen Zhen Zhao; Kelly Iyadurai; Anjali K. Henders; Nils Homer; Megan J. Campbell; Mitchell S. Stark; Shane Thomas; Helen Schmid; Elizabeth A. Holland; Elizabeth M. Gillanders; David L. Duffy; Judith A. Maskiell; Jodie Jetann; Megan Ferguson; Dietrich A. Stephan; Anne E. Cust; David C. Whiteman; Adèle C. Green; Håkan Olsson; Susana Puig; Paola Ghiorzo; Johan Hansson; Florence Demenais; Alisa M. Goldstein; Nelleke A. Gruis; David E. Elder

We conducted a genome-wide association pooling study for cutaneous melanoma and performed validation in samples totaling 2,019 cases and 2,105 controls. Using pooling, we identified a new melanoma risk locus on chromosome 20 (rs910873 and rs1885120), with replication in two further samples (combined P < 1 × 10−15). The per allele odds ratio was 1.75 (1.53, 2.01), with evidence for stronger association in early-onset cases.


Nature Genetics | 2011

Genome-wide association study identifies three new melanoma susceptibility loci

Jennifer H. Barrett; Mark M. Iles; Mark Harland; John C. Taylor; Joanne F. Aitken; Per Arne Andresen; Lars A. Akslen; Bruce K. Armstrong; M.-F. Avril; Esther Azizi; Bert Bakker; Wilma Bergman; Giovanna Bianchi-Scarrà; Brigitte Bressac-de Paillerets; Donato Calista; Lisa A. Cannon-Albright; Eve Corda; Anne E. Cust; Tadeusz Dȩbniak; David L. Duffy; Alison M. Dunning; Douglas F. Easton; Eitan Friedman; Pilar Galan; Paola Ghiorzo; Graham G. Giles; Johan Hansson; Marko Hocevar; Veronica Höiom; John L. Hopper

We report a genome-wide association study for melanoma that was conducted by the GenoMEL Consortium. Our discovery phase included 2,981 individuals with melanoma and 1,982 study-specific control individuals of European ancestry, as well as an additional 6,426 control subjects from French or British populations, all of whom were genotyped for 317,000 or 610,000 single-nucleotide polymorphisms (SNPs). Our analysis replicated previously known melanoma susceptibility loci. Seven new regions with at least one SNP with P < 10−5 and further local imputed or genotyped support were selected for replication using two other genome-wide studies (from Australia and Texas, USA). Additional replication came from case-control series from the UK and The Netherlands. Variants at three of the seven loci replicated at P < 10−3: an SNP in ATM (rs1801516, overall P = 3.4 × 10−9), an SNP in MX2 (rs45430, P = 2.9 × 10−9) and an SNP adjacent to CASP8 (rs13016963, P = 8.6 × 10−10). A fourth locus near CCND1 remains of potential interest, showing suggestive but inconclusive evidence of replication (rs1485993, overall P = 4.6 × 10−7 under a fixed-effects model and P = 1.2 × 10−3 under a random-effects model). These newly associated variants showed no association with nevus or pigmentation phenotypes in a large British case-control series.


Nature Genetics | 2014

Rare missense variants in POT1 predispose to familial cutaneous malignant melanoma

Jianxin Shi; Xiaohong R. Yang; Bari J. Ballew; Melissa Rotunno; Donato Calista; Maria Concetta Fargnoli; Paola Ghiorzo; Brigitte Bressac-de Paillerets; Eduardo Nagore; M.-F. Avril; Neil E. Caporaso; Mary L. McMaster; Michael Cullen; Zhaoming Wang; Xijun Zhang; William Bruno; Lorenza Pastorino; Paola Queirolo; Jose Banuls-Roca; Zaida García-Casado; Amaury Vaysse; Hamida Mohamdi; Yasser Riazalhosseini; Mario Foglio; Fanélie Jouenne; Xing Hua; Paula L. Hyland; Jinhu Yin; Haritha Vallabhaneni; Weihang Chai

Although CDKN2A is the most frequent high-risk melanoma susceptibility gene, the underlying genetic factors for most melanoma-prone families remain unknown. Using whole-exome sequencing, we identified a rare variant that arose as a founder mutation in the telomere shelterin gene POT1 (chromosome 7, g.124493086C>T; p.Ser270Asn) in five unrelated melanoma-prone families from Romagna, Italy. Carriers of this variant had increased telomere lengths and numbers of fragile telomeres, suggesting that this variant perturbs telomere maintenance. Two additional rare POT1 variants were identified in all cases sequenced in two separate Italian families, one variant per family, yielding a frequency for POT1 variants comparable to that for CDKN2A mutations in this population. These variants were not found in public databases or in 2,038 genotyped Italian controls. We also identified two rare recurrent POT1 variants in US and French familial melanoma cases. Our findings suggest that POT1 is a major susceptibility gene for familial melanoma in several populations.


American Journal of Medical Genetics Part A | 2009

Identification of a SUFU germline mutation in a family with Gorlin syndrome.

Lorenza Pastorino; Paola Ghiorzo; Sabina Nasti; Roberto Cusano; C. Marzocchi; Ml Garre; M. Clementi; G. Bianchi Scarrà

Gorlin syndrome (GS) is inherited in an autosomal dominant pattern with high‐penetrance and is characterized by a range of developmental anomalies and increased risk of developing basal cell carcinoma and medulloblastoma. Between 50% and 85% of patients with GS harbor germ line mutations in the only susceptibility gene identified to date, PTCH1, a key component in the Sonic Hedgehog signaling pathway. Another component in this pathway, SUFU, is known to be involved in susceptibility to medulloblastoma but has never been reported in GS patients to date. We have identified the known c.1022 + 1G>A SUFU germ line splicing mutation in a family that was PTCH1‐negative and who had signs and symptoms of GS, including medulloblastoma. This is the first report of a germ line SUFU mutation associated with GS.


International Journal of Cancer | 1999

Characterization of ligurian melanoma families and risk of occurrence of other neoplasia

Paola Ghiorzo; Paola Ciotti; Michela Mantelli; Abdelhamid Heouaine; Paola Queirolo; Maria Luisa Rainero; Carlo Ferrari; Pier Luigi Santi; Roberto De Marchi; A. Farris; F. Ajmar; Paolo Bruzzi; Giovanna Bianchi-Scarrà

Germline mutations impairing the p16INK4‐function have previously been demonstrated to be responsible for genetic predisposition in at least one half of melanoma‐prone kindreds of North European origin. Familial melanoma kindreds have also been found to present an increased risk of pancreatic cancer and other cancers, but results relative to more common neoplasias incidence, in particular, are heterogeneous. We report here a clinical‐epidemiological study, including the presence of additional neoplasias, in 14 apparently unrelated kindreds coming from a small geographic region of Northern Italy (Liguria), having therefore lived for generations in similar environmental conditions. We identified the common p16 missense mutation (Gly101Trp) reported in several previously studied kindreds, in 7 of 14 families, whereas the remaining 7 families had no detectable mutations in the coding region of p16 gene. Median age at diagnosis and other melanoma features were studied. When compared with the expected figures, based on regional incidence rates, a significant excess of pancreatic cancer, with 4 cases diagnosed, and of breast cancer, with 7 cases, was observed. The 7 families without apparent CDKN2A involvement were also negative for hot‐spot exon 2 mutation of CDK4. Environmental factors do not appear to play a role in the excess of non‐melanoma neoplasia in our families, as somewhat substantiated by the control group, composed of spouses and members of non‐affected branches; they do not reveal any increased cancer incidence compared with the general population. Furthermore, given the proven significance of interaction between the melanoma susceptibility gene and the propensity to sunburns and other environmental risk factors, our results, obtained from a small but homogeneous sample, may have important implications for further risk assessment studies. Int. J. Cancer 83:441–448, 1999.


Nature Genetics | 2015

Genome-wide meta-analysis identifies five new susceptibility loci for cutaneous malignant melanoma

Matthew H. Law; D. Timothy Bishop; Jeffrey E. Lee; Myriam Brossard; Nicholas G. Martin; Eric K. Moses; Fengju Song; Jennifer H. Barrett; Rajiv Kumar; Douglas F. Easton; Paul Pharoah; Anthony J. Swerdlow; Katerina P. Kypreou; John C. Taylor; Mark Harland; Juliette Randerson-Moor; Lars A. Akslen; Per Arne Andresen; M.-F. Avril; Esther Azizi; Giovanna Bianchi Scarrà; Kevin M. Brown; Tadeusz Dębniak; David L. Duffy; David E. Elder; Shenying Fang; Eitan Friedman; Pilar Galan; Paola Ghiorzo; Elizabeth M. Gillanders

Thirteen common susceptibility loci have been reproducibly associated with cutaneous malignant melanoma (CMM). We report the results of an international 2-stage meta-analysis of CMM genome-wide association studies (GWAS). This meta-analysis combines 11 GWAS (5 previously unpublished) and a further three stage 2 data sets, totaling 15,990 CMM cases and 26,409 controls. Five loci not previously associated with CMM risk reached genome-wide significance (P < 5 × 10−8), as did 2 previously reported but unreplicated loci and all 13 established loci. Newly associated SNPs fall within putative melanocyte regulatory elements, and bioinformatic and expression quantitative trait locus (eQTL) data highlight candidate genes in the associated regions, including one involved in telomere biology.

Collaboration


Dive into the Paola Ghiorzo's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Paola Queirolo

National Cancer Research Institute

View shared research outputs
Top Co-Authors

Avatar

Maria Teresa Landi

United States Department of Health and Human Services

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Alisa M. Goldstein

National Institutes of Health

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicholas K. Hayward

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar

Donato Calista

Pomeranian Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge