Network


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

Hotspot


Dive into the research topics where Marta Betti is active.

Publication


Featured researches published by Marta Betti.


Genes, Chromosomes and Cancer | 2015

Inference on germline BAP1 mutations and asbestos exposure from the analysis of familial and sporadic mesothelioma in a high‐risk area

Marta Betti; Elisabetta Casalone; Daniela Ferrante; Antonio Romanelli; Federica Grosso; Simonetta Guarrera; Luisella Righi; Simona Vatrano; Giuseppe Pelosi; Roberta Libener; Dario Mirabelli; Renzo Boldorini; Caterina Casadio; Mauro Papotti; Giuseppe Matullo; Corrado Magnani; Irma Dianzani

Inherited loss‐of‐function mutations in the BAP1 oncosuppressor gene are responsible for an inherited syndrome with predisposition to malignant mesothelioma (MM), uveal and keratinocytic melanoma, and other malignancies. Germline mutations that were inherited in an autosomal dominant fashion were identified in nine families with multiplex MM cases and 25 families with multiple melanoma, renal cell carcinoma, and other tumors. Germline mutations were also identified in sporadic MM cases, suggesting that germline mutations in BAP1 occur frequently. In this article, we report the analysis of BAP1 in five multiplex MM families and in 103 sporadic cases of MM. One family carried a new truncating germline mutation. Using immunohistochemistry, we show that BAP1 is not expressed in tumor tissue, which is in accordance with Knudsons two hits hypothesis. Interestingly, whereas the three individuals who were possibly exposed to asbestos developed MM, the individual who was not exposed developed a different tumor type, that is, mucoepidermoid carcinoma. This finding suggests that the type of carcinogen exposure may be important for the cancer type that is developed by mutation carriers. On the contrary, the other families or the 103 sporadic patients did not show germline mutations in BAP1. Our data show that BAP1 mutations are very rare in patients with sporadic MM, and we report a new BAP1 mutation, extend the cancer types associated with these mutations, and suggest the existence of other yet unknown genes in the pathogenesis of familial MM.


PLOS ONE | 2012

An Overview of the Genetic Structure within the Italian Population from Genome-Wide Data

Cornelia Di Gaetano; Floriana Voglino; Simonetta Guarrera; Giovanni Fiorito; Fabio Rosa; Anna Maria Di Blasio; Paola Manzini; Irma Dianzani; Marta Betti; Daniele Cusi; Francesca Frau; Cristina Barlassina; Dario Mirabelli; Corrado Magnani; Nicola Glorioso; Stefano Bonassi; Alberto Piazza; Giuseppe Matullo

In spite of the common belief of Europe as reasonably homogeneous at genetic level, advances in high-throughput genotyping technology have resolved several gradients which define different geographical areas with good precision. When Northern and Southern European groups were considered separately, there were clear genetic distinctions. Intra-country genetic differences were also evident, especially in Finland and, to a lesser extent, within other European populations. Here, we present the first analysis using the 125,799 genome-wide Single Nucleotide Polymorphisms (SNPs) data of 1,014 Italians with wide geographical coverage. We showed by using Principal Component analysis and model-based individual ancestry analysis, that the current population of Sardinia can be clearly differentiated genetically from mainland Italy and Sicily, and that a certain degree of genetic differentiation is detectable within the current Italian peninsula population. Pair-wise FST statistics Northern and Southern Italy amounts approximately to 0.001 between, and around 0.002 between Northern Italy and Utah residents with Northern and Western European ancestry (CEU). The Italian population also revealed a fine genetic substructure underscoring by the genomic inflation (Sardinia vs. Northern Italy = 3.040 and Northern Italy vs. CEU = 1.427), warning against confounding effects of hidden relatedness and population substructure in association studies.


Mutation Research | 2011

XRCC1 and ERCC1 variants modify malignant mesothelioma risk: A case–control study

Marta Betti; Daniela Ferrante; Marina Padoan; Simonetta Guarrera; Mara Giordano; Anna Aspesi; Dario Mirabelli; Caterina Casadio; Francesco Ardissone; Enrico Ruffini; Pier Giacomo Betta; Roberta Libener; Roberto Guaschino; Giuseppe Matullo; E. Piccolini; Corrado Magnani; Irma Dianzani

Malignant pleural mesothelioma (MPM) is a rare aggressive tumor associated with asbestos exposure. The possible role of genetic factors has also been suggested and MPM has been associated with single nucleotide polymorphisms (SNPs) of xenobiotic and oxidative metabolism enzymes. We have identified an association of the DNA repair gene XRCC1 with MPM in the population of Casale Monferrato, a town exposed to high asbestos pollution. To extend this observation we examined 35 SNPs in 15 genes that could be involved in MPM carcinogenicity in 220 MPM patients and 296 controls from two case-control studies conducted in Casale (151 patients, 252 controls) and Turin (69 patients, 44 controls), respectively. Unconditional multivariate logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (95% CIs). Two DNA repair genes were associated with MPM, i.e. XRCC1 and ERCC1. Considering asbestos-exposed only, the risk increased with the increasing number of XRCC1-399Q alleles (Casale: OR=1.44, 95%CI 1.02-2.03; Casale+Turin: OR=1.34, 95%CI 0.98-1.84) or XRCC1 -77T alleles (Casale+Turin: OR=1.33, 95%CI 0.97-1.81). The XRCC1-TGGGGGAACAGA haplotype was significantly associated with MPM (Casale: OR=1.76, 95%CI 1.04-2.96). Patients heterozygotes for ERCC1 N118N showed an increased OR in all subjects (OR=1.66, 95%CI 1.06-2.60) and in asbestos-exposed only (OR=1.59, 95%CI 1.01-2.50). When the dominant model was considered (i.e. ERCC1 heterozygotes CT plus homozygotes CC versus homozygotes TT) the risk was statistically significant both in all subjects (OR=1.61, 95%CI 1.06-2.47) and in asbestos-exposed only (OR=1.56, 95%CI 1.02-2.40). The combination of ERCC1 N118N and XRCC1 R399Q was statistically significant (Casale: OR=2.02, 95%CI 1.01-4.05; Casale+Turin: OR=2.39, 95%CI 1.29-4.43). The association of MPM with DNA repair genes support the hypothesis that an increased susceptibility to DNA damage may favour asbestos carcinogenicity.


PLOS ONE | 2013

Genetic Variants Associated with Increased Risk of Malignant Pleural Mesothelioma: A Genome-Wide Association Study

Giuseppe Matullo; Simonetta Guarrera; Marta Betti; Giovanni Fiorito; Daniela Ferrante; Floriana Voglino; Gemma Cadby; Cornelia Di Gaetano; Fabio Rosa; Alessia Russo; Ari Hirvonen; Elisabetta Casalone; Sara Tunesi; Marina Padoan; Mara Giordano; Anna Aspesi; Caterina Casadio; Francesco Ardissone; Enrico Ruffini; Pier Giacomo Betta; Roberta Libener; Roberto Guaschino; Ezio Piccolini; Monica Neri; Arthur W. Musk; Nicholas de Klerk; Jennie Hui; John Beilby; Alan James; Jenette Creaney

Asbestos exposure is the main risk factor for malignant pleural mesothelioma (MPM), a rare aggressive tumor. Nevertheless, only 5–17% of those exposed to asbestos develop MPM, suggesting the involvement of other environmental and genetic risk factors. To identify the genetic risk factors that may contribute to the development of MPM, we conducted a genome-wide association study (GWAS; 370,000 genotyped SNPs, 5 million imputed SNPs) in Italy, among 407 MPM cases and 389 controls with a complete history of asbestos exposure. A replication study was also undertaken and included 428 MPM cases and 1269 controls from Australia. Although no single marker reached the genome-wide significance threshold, several associations were supported by haplotype-, chromosomal region-, gene- and gene-ontology process-based analyses. Most of these SNPs were located in regions reported to harbor aberrant alterations in mesothelioma (SLC7A14, THRB, CEBP350, ADAMTS2, ETV1, PVT1 and MMP14 genes), causing at most a 2–3-fold increase in MPM risk. The Australian replication study showed significant associations in five of these chromosomal regions (3q26.2, 4q32.1, 7p22.2, 14q11.2, 15q14). Multivariate analysis suggested an independent contribution of 10 genetic variants, with an Area Under the ROC Curve (AUC) of 0.76 when only exposure and covariates were included in the model, and of 0.86 when the genetic component was also included, with a substantial increase of asbestos exposure risk estimation (odds ratio, OR: 45.28, 95% confidence interval, CI: 21.52–95.28). These results showed that genetic risk factors may play an additional role in the development of MPM, and that these should be taken into account to better estimate individual MPM risk in individuals who have been exposed to asbestos.


Cancer Letters | 2016

CDKN2A and BAP1 germline mutations predispose to melanoma and mesothelioma

Marta Betti; Anna Aspesi; Alessandra Biasi; Elisabetta Casalone; Daniela Ferrante; Paola Ogliara; Laura Cristina Gironi; Roberto Giorgione; Pamela Farinelli; Federica Grosso; Roberta Libener; S. Rosato; Daniela Turchetti; Antonella Maffè; Caterina Casadio; Valeria Ascoli; Chiara Dianzani; Enrico Colombo; E. Piccolini; Mansueto Pavesi; Sara Miccoli; Dario Mirabelli; Cecilia Bracco; Luisella Righi; Renzo Boldorini; Mauro Papotti; Giuseppe Matullo; Corrado Magnani; Barbara Pasini; Irma Dianzani

BAP1 germline mutations predispose to a cancer predisposition syndrome that includes mesothelioma, cutaneous melanoma, uveal melanoma and other cancers. This co-occurrence suggests that these tumors share a common carcinogenic pathway. To evaluate this hypothesis, we studied 40 Italian families with mesothelioma and/or melanoma. The probands were sequenced for BAP1 and for the most common melanoma predisposition genes (i.e. CDKN2A, CDK4, TERT, MITF and POT1) to investigate if these genes may also confer susceptibility to mesothelioma. In two out of six families with both mesothelioma and melanoma we identified either a germline nonsense mutation (c.1153C > T, p.Arg385*) in BAP1 or a recurrent pathogenic germline mutation (c.301G > T, p.Gly101Trp) in CDKN2A. Our study suggests that CDKN2A, in addition to BAP1, could be involved in the melanoma and mesothelioma susceptibility, leading to the rare familial cancer syndromes. It also suggests that these tumors share key steps that drive carcinogenesis and that other genes may be involved in inherited predisposition to malignant mesothelioma and melanoma.


International Journal of Hygiene and Environmental Health | 2009

Pooled analysis of NAT2 genotypes as risk factors for asbestos-related malignant mesothelioma

Marta Betti; Monica Neri; Daniela Ferrante; Stefano Landi; Alessandra Biava; Federica Gemignani; Marinella Bertolotti; Dario Mirabelli; Marina Padoan; Donatella Ugolini; Mario Botta; Stefano Bonassi; Corrado Magnani; Irma Dianzani

Malignant mesothelioma (MM) is a rare and aggressive tumor of the pleura. The most important causal factor for the development of MM is occupational exposure to asbestos. Different lines of evidence suggest a role of genetic background in MM development, as for other cancers. Two published studies observed an association between MM and N-acetyl-transferase 2 (NAT2) polymorphisms. First, a Finnish study observed that the NAT2 slow acetylator phenotype was associated with an increased risk of MM. Conversely, MM risk was higher in Italian subjects carrying the NAT2 fast acetylator genotypes. The conflicting results obtained in Finland and Italy could be ascribed to random chance, considering the small panel of patients and controls in the two studies, but also ethnic or other differences may have been important. To ascertain the role of NAT2 genotype, we performed a study on 252 MM patients and 262 controls recruited in two Northern Italy areas that were characterized by high asbestos exposure, due to intense industrial activities (an asbestos cement factory in Casale Monferrato, mainly shipyards and refineries in Liguria). Unconditional multivariate logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs). NAT2 fast acetylator genotypes showed an increased OR, although not statistically significant, both in asbestos-exposed subjects (OR=1.47; 95% CI=0.96-2.26) and in the entire population (OR=1.38; 95% CI=0.93-2.04). These results suggest that NAT2 polymorphisms do not exert a strong effect on individual susceptibility to MM.


Carcinogenesis | 2015

Gene-asbestos interaction in malignant pleural mesothelioma susceptibility

Sara Tunesi; Daniela Ferrante; Dario Mirabelli; S. Andorno; Marta Betti; Giovanni Fiorito; Simonetta Guarrera; Elisabetta Casalone; Monica Neri; Donatella Ugolini; Stefano Bonassi; Giuseppe Matullo; Irma Dianzani; Corrado Magnani

Asbestos exposure is the main risk factor for malignant pleural mesothelioma (MPM), a rare aggressive tumor. Nevertheless, on average less than 10% of subjects highly exposed to asbestos develop MPM, suggesting the possible involvement of other risk factors. To identify the genetic factors that may modulate the risk of MPM, we conducted a gene-environment interaction analysis including asbestos exposure and 15 single nucleotide polymorphisms (SNPs) previously identified through a genome-wide association study on Italian subjects. In the present study, we assessed gene-asbestos interaction on MPM risk using relative excess risk due to interaction and synergy index for additive interaction and V index for multiplicative interaction. Generalized multifactor dimensionality reduction (GMDR) analyses were also performed. Positive deviation from additivity was found for six SNPs (rs1508805, rs2501618, rs4701085, rs4290865, rs10519201, rs763271), and four of them (rs1508805, rs2501618, rs4701085, rs10519201) deviated also from multiplicative models. However, after Bonferroni correction, deviation from multiplicative model was still significant for rs1508805 and rs4701085 only. GMDR analysis showed a strong MPM risk due to asbestos exposure and suggested a possible synergistic effect between asbestos exposure and rs1508805, rs2501618 and rs5756444. Our results suggested that gene-asbestos interaction may play an additional role on MPM susceptibility, given that asbestos exposure appears as the main risk factor.


Scientific Reports | 2017

Lymphoblastoid cell lines from Diamond Blackfan anaemia patients exhibit a full ribosomal stress phenotype that is rescued by gene therapy

Anna Aspesi; Valentina Monteleone; Marta Betti; Chiara Actis; Giulia Morleo; Marika Sculco; Simonetta Guarrera; Marcin W. Wlodarski; Ugo Ramenghi; Claudio Santoro; Steven R. Ellis; Fabrizio Loreni; Antonia Follenzi; Irma Dianzani

Diamond Blackfan anaemia (DBA) is a congenital bone marrow failure syndrome characterised by selective red cell hypoplasia. DBA is most often due to heterozygous mutations in ribosomal protein (RP) genes that lead to defects in ribosome biogenesis and function and result in ribosomal stress and p53 activation. The molecular mechanisms underlying this pathology are still poorly understood and studies on patient erythroid cells are hampered by their paucity. Here we report that RP-mutated lymphoblastoid cell lines (LCLs) established from DBA patients show defective rRNA processing and ribosomal stress features such as reduced proliferation, decreased protein synthesis, and activation of p53 and its target p21. These phenotypic alterations were corrected by gene complementation. Our data indicate that DBA LCLs could be a useful model for molecular and pharmacological investigations.


PLOS ONE | 2015

Erratum: Genetic variants associated with increased risk of malignant pleural mesothelioma: A genome-wide association study (PLoS ONE (2015) 8:4 (e61253) DOI: 10.1371/journal.pone.0061253)

Giuseppe Matullo; Simonetta Guarrera; Marta Betti; Giovanni Fiorito; Daniela Ferrante; Floriana Voglino; Gemma Cadby; Cornelia Di Gaetano; Fabio Rosa; Alessia Russo; Ari Hirvonen; Elisabetta Casalone; Sara Tunesi; Marina Padoan; Mara Giordano; Anna Aspesi; Caterina Casadio; Francesco Ardissone; Enrico Ruffini; Pier Giacomo Betta; Roberta Libener; Roberto Guaschino; Ezio Piccolini; Monica Neri; Arthur W. Musk; Nicholas de Klerk; Jennie Hui; John Beilby; Alan James; Jenette Creaney

Giuseppe Matullo, Simonetta Guarrera, Marta Betti, Giovanni Fiorito, Daniela Ferrante, Floriana Voglino, Gemma Cadby, Cornelia Di Gaetano, Fabio Rosa, Alessia Russo, Ari Hirvonen, Elisabetta Casalone, Sara Tunesi, Marina Padoan, Mara Giordano, Anna Aspesi, Caterina Casadio, Francesco Ardissone, Enrico Ruffini, Pier Giacomo Betta, Roberta Libener, Roberto Guaschino, Ezio Piccolini, Monica Neri, Arthur W. B. Musk, Nicholas H. de Klerk, Jennie Hui, John Beilby, Alan L. James, Jenette Creaney, Bruce W. Robinson, Sutapa Mukherjee, Lyle J. Palmer, Dario Mirabelli, Donatella Ugolini, Stefano Bonassi, Corrado Magnani, Irma Dianzani


Human Mutation | 2018

A functional assay for the clinical annotation of genetic variants of uncertain significance in Diamond-Blackfan anemia

Anna Aspesi; Marta Betti; Marika Sculco; Chiara Actis; Cristina Olgasi; Marcin W. Wlodarski; Adrianna Vlachos; Jeffrey M. Lipton; Ugo Ramenghi; Claudio Santoro; Antonia Follenzi; Steven R. Ellis; Irma Dianzani

Diamond–Blackfan anemia (DBA) is a rare genetic hypoplasia of erythroid progenitors characterized by mild to severe anemia and associated with congenital malformations. Clinical manifestations in DBA patients are quite variable and genetic testing has become a critical factor in establishing a diagnosis of DBA. The majority of DBA cases are due to heterozygous loss‐of‐function mutations in ribosomal protein (RP) genes. Causative mutations are fairly straightforward to identify in the case of large deletions and frameshift and nonsense mutations found early in a protein coding sequence, but diagnosis becomes more challenging in the case of missense mutations and small in‐frame indels. Our group recently characterized the phenotype of lymphoblastoid cell lines established from DBA patients with pathogenic lesions in RPS19 and observed that defective pre‐rRNA processing, a hallmark of the disease, was rescued by lentiviral vectors expressing wild‐type RPS19. Here, we use this complementation assay to determine whether RPS19 variants of unknown significance are capable of rescuing pre‐rRNA processing defects in these lymphoblastoid cells as a means of assessing the effects of these sequence changes on the function of the RPS19 protein. This approach will be useful in differentiating pathogenic mutations from benign polymorphisms in identifying causative genes in DBA patients.

Collaboration


Dive into the Marta Betti's collaboration.

Top Co-Authors

Avatar

Irma Dianzani

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniela Ferrante

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Corrado Magnani

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Anna Aspesi

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Caterina Casadio

University of Eastern Piedmont

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge