1
|
Yeh I. Melanocytic naevi, melanocytomas and emerging concepts. Pathology 2023; 55:178-186. [PMID: 36642570 DOI: 10.1016/j.pathol.2022.12.341] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 12/15/2022] [Accepted: 12/19/2022] [Indexed: 12/28/2022]
Abstract
With the elucidation of the genetics of melanocytic tumours, new concepts have emerged. An important one is the identification of 'intermediate' melanocytic tumours, those with genetic progression events beyond those of melanocytic naevi but that are not fully malignant. Thus, melanocytic tumours exist on a genetic spectrum that likely corresponds to biological behaviour. There are multiple pathways to melanoma development with different initiating events and characteristic benign melanocytic neoplasms and the precise placement of tumours on these pathways remains to be established and the corresponding risks of progression quantified. In this review, I discuss the classification of melanocytic naevi based on clinical, histopathological and genetic features, as well as the concept of melanocytomas with discussion of specific recognisable subtypes.
Collapse
Affiliation(s)
- Iwei Yeh
- Departments of Dermatology and Pathology, University of California, San Francisco, CA, USA.
| |
Collapse
|
2
|
The Complex Interplay between Nevi and Melanoma: Risk Factors and Precursors. Int J Mol Sci 2023; 24:ijms24043541. [PMID: 36834954 PMCID: PMC9964821 DOI: 10.3390/ijms24043541] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2022] [Revised: 01/30/2023] [Accepted: 02/07/2023] [Indexed: 02/12/2023] Open
Abstract
One effort to combat the rising incidence of malignant melanoma is focused on early detection by the clinical and dermoscopic screening of melanocytic nevi. However, the interaction between nevi, which are congenital or acquired benign melanocytic proliferations, and melanoma is still enigmatic. On the one hand, the majority of melanomas are thought to form de novo, as only a third of primary melanomas are associated with a histologically identifiable nevus precursor. On the other hand, an increased number of melanocytic nevi is a strong risk factor for developing melanoma, including melanomas that do not derive from nevi. The formation of nevi is modulated by diverse factors, including pigmentation, genetic risk factors, and environmental sun exposure. While the molecular alterations that occur during the progression of a nevus to melanoma have been well characterized, many unanswered questions remain surrounding the process of nevus to melanoma evolution. In this review, we discuss clinical, histological, molecular, and genetic factors that influence nevus formation and progression to melanoma.
Collapse
|
3
|
Calbet‐Llopart N, Combalia M, Kiroglu A, Potrony M, Tell‐Martí G, Combalia A, Brugues A, Podlipnik S, Carrera C, Puig S, Malvehy J, Puig‐Butillé JA. Common genetic variants associated with melanoma risk or naevus count in patients with wildtype MC1R melanoma. Br J Dermatol 2022; 187:753-764. [PMID: 35701387 PMCID: PMC9804579 DOI: 10.1111/bjd.21707] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 06/07/2022] [Accepted: 06/11/2022] [Indexed: 01/05/2023]
Abstract
BACKGROUND Hypomorphic MC1R variants are the most prevalent genetic determinants of melanoma risk in the white population. However, the genetic background of patients with wildtype (WT) MC1R melanoma is poorly studied. OBJECTIVES To analyse the role of candidate common genetic variants on the melanoma risk and naevus count in Spanish patients with WT MC1R melanoma. METHODS We examined 753 individuals with WT MC1R from Spain (497 patients and 256 controls). We used OpenArray reverse-transcriptase polymerase chain reaction to genotype a panel of 221 common genetic variants involved in melanoma, naevogenesis, hormonal pathways and proinflammatory pathways. Genetic models were tested using multivariate logistic regression models. Nonparametric multifactor dimensionality reduction (MDR) was used to detect gene-gene interactions within each biological subgroup of variants. RESULTS We found that variant rs12913832 in the HERC2 gene, which is associated with blue eye colour, increased melanoma risk in individuals with WT MC1R [odds ratio (OR) 1·97, 95% confidence interval (CI) 1·48-2·63; adjusted P < 0·001; corrected P < 0·001]. We also observed a trend between the rs3798577 variant in the oestrogen receptor alpha gene (ESR1) and a lower naevus count, which was restricted to female patients with WT MC1R (OR 0·51, 95% CI 0·33-0·79; adjusted P = 0·002; corrected P = 0·11). This sex-dependent association was statistically significant in a larger cohort of patients with melanoma regardless of their MC1R status (n = 1497; OR 0·71, 95% CI 0·57-0·88; adjusted P = 0·002), reinforcing the hypothesis of an association between hormonal pathways and susceptibility to melanocytic proliferation. Last, the MDR analysis revealed four genetic combinations associated with melanoma risk or naevus count in patients with WT MC1R. CONCLUSIONS Our data suggest that epistatic interaction among common variants related to melanocyte biology or proinflammatory pathways might influence melanocytic proliferation in individuals with WT MC1R. What is already known about this topic? Genetic variants in the MC1R gene are the most prevalent melanoma genetic risk factor in the white population. Still, 20-40% of cases of melanoma occur in individuals with wildtype MC1R. Multiple genetic variants have a pleiotropic effect in melanoma and naevogenesis. Additional variants in unexplored pathways might also have a role in melanocytic proliferation in these patients. Epidemiological evidence suggests an association of melanocytic proliferation with hormonal pathways and proinflammatory pathways. What does this study add? Variant rs12913832 in the HERC2 gene, which is associated with blue eye colour, increases the melanoma risk in individuals with wildtype MC1R. Variant rs3798577 in the oestrogen receptor gene is associated with naevus count regardless of the MC1R status in female patients with melanoma. We report epistatic interactions among common genetic variants with a role in modulating the risk of melanoma or the number of naevi in individuals with wildtype MC1R. What is the translational message? We report a potential role of hormonal signalling pathways in melanocytic proliferation, providing a basis for better understanding of sex-based differences observed at the epidemiological level. We show that gene-gene interactions among common genetic variants might be responsible for an increased risk for melanoma development in individuals with a low-risk phenotype, such as darkly pigmented hair and skin.
Collapse
Affiliation(s)
- Neus Calbet‐Llopart
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIBarcelonaSpain
| | - Marc Combalia
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Anil Kiroglu
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Miriam Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIBarcelonaSpain,Biochemistry and Molecular Genetics DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Gemma Tell‐Martí
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIBarcelonaSpain
| | - Andrea Combalia
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Albert Brugues
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Sebastian Podlipnik
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain
| | - Cristina Carrera
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIBarcelonaSpain
| | - Susana Puig
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIBarcelonaSpain
| | - Josep Malvehy
- Dermatology DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIBarcelonaSpain
| | - Joan Anton Puig‐Butillé
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER)Instituto de Salud Carlos IIIBarcelonaSpain,Molecular Biology CORE, Biochemistry and Molecular Genetics DepartmentMelanoma Group, Hospital Clínic de Barcelona, IDIBAPS, University of BarcelonaBarcelonaSpain
| |
Collapse
|
4
|
Wang Y, Song H, Miao Q, Wang Y, Qi J, Xu X, Sun J. PLA2G6 Silencing Suppresses Melanoma Progression and Affects Ferroptosis Revealed by Quantitative Proteomics. Front Oncol 2022; 12:819235. [PMID: 35340268 PMCID: PMC8948425 DOI: 10.3389/fonc.2022.819235] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Accepted: 02/04/2022] [Indexed: 12/28/2022] Open
Abstract
Although phospholipase A2 group VI (PLA2G6) is involved in oncogenesis in several human tumors, its expression and role in cutaneous malignant melanoma (CMM) pathogenesis remains unclear. Here, by using the Oncomine and CCLE online database, immunohistochemistry, RT-qPCR, and western blotting analysis, we revealed that PLA2G6 was markedly up-regulated in CMM tissues compared to nevus tissues, as well as remarkably increased in vitro in SK-MEL-28 and M14 melanoma cell lines compared to human melanocytes. In vivo, PLA2G6 was also elevated in nine melanoma tissues compared to adjacent tissues. To investigate the malignant behaviors of PLA2G6 in CMM, SK-MEL-28 and M14 cell lines with PLA2G6 stable knockdown by RNAi strategy were constructed. Through CCK8 and colony formation assays in vitro and xenograft tumor experiment in vivo, we found that knockdown of PLA2G6 dramatically inhibited cell proliferation. The results of scratch-wound and transwell assays suggested that the migration and invasion of melanoma cells were prominently suppressed after silencing PLA2G6. In addition, flow cytometry showed that the knockdown of PLA2G6 promoted the apoptosis rate of melanoma cells. To further explore the potential molecular mechanism, we used liquid chromatography coupled to tandem mass spectrometry (LC-MS/MS) proteomic and bioinformatics analysis. The GO and KEGG analysis suggested that the underlying mechanism of PLA2G6 in CMM might be associated with the ferroptosis pathway, and ferroptosis-related proteins were validated to be differentially expressed in PLA2G6 knockdown SK-MEL-28 and M14 cells. Together, these results suggested that PLA2G6 knockdown significantly inhibited cell proliferation, metastasis, and promoted apoptosis in melanoma. Our findings on the biological function of PLA2G6 and the underlying association between PLA2G6 and ferroptosis in melanoma may contribute to developing a potential therapeutic strategy for melanoma.
Collapse
Affiliation(s)
- Yifei Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Hao Song
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Qiuju Miao
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Yan Wang
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jinliang Qi
- State Key Laboratory of Pharmaceutical Biotechnology, School of Life Sciences, Nanjing University, Nanjing, China
| | - Xiulian Xu
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| | - Jianfang Sun
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, China
| |
Collapse
|
5
|
Association of Melanoma-Risk Variants with Primary Melanoma Tumor Prognostic Characteristics and Melanoma-Specific Survival in the GEM Study. Curr Oncol 2021; 28:4756-4771. [PMID: 34898573 PMCID: PMC8628692 DOI: 10.3390/curroncol28060401] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 10/11/2021] [Accepted: 11/10/2021] [Indexed: 12/15/2022] Open
Abstract
Genome-wide association studies (GWAS) and candidate pathway studies have identified low-penetrant genetic variants associated with cutaneous melanoma. We investigated the association of melanoma-risk variants with primary melanoma tumor prognostic characteristics and melanoma-specific survival. The Genes, Environment, and Melanoma Study enrolled 3285 European origin participants with incident invasive primary melanoma. For each of 47 melanoma-risk single nucleotide polymorphisms (SNPs), we used linear and logistic regression modeling to estimate, respectively, the per allele mean changes in log of Breslow thickness and odds ratios for presence of ulceration, mitoses, and tumor-infiltrating lymphocytes (TILs). We also used Cox proportional hazards regression modeling to estimate the per allele hazard ratios for melanoma-specific survival. Passing the false discovery threshold (p = 0.0026) were associations of IRF4 rs12203592 and CCND1 rs1485993 with log of Breslow thickness, and association of TERT rs2242652 with presence of mitoses. IRF4 rs12203592 also had nominal associations (p < 0.05) with presence of mitoses and melanoma-specific survival, as well as a borderline association (p = 0.07) with ulceration. CCND1 rs1485993 also had a borderline association with presence of mitoses (p = 0.06). MX2 rs45430 had nominal associations with log of Breslow thickness, presence of mitoses, and melanoma-specific survival. Our study indicates that further research investigating the associations of these genetic variants with underlying biologic pathways related to tumor progression is warranted.
Collapse
|
6
|
Xu M, Mehl L, Zhang T, Thakur R, Sowards H, Myers T, Jessop L, Chesi A, Johnson ME, Wells AD, Michael HT, Bunda P, Jones K, Higson H, Hennessey RC, Jermusyk A, Kovacs MA, Landi MT, Iles MM, Goldstein AM, Choi J, Chanock SJ, Grant SF, Chari R, Merlino G, Law MH, Brown KM, Brown KM. A UVB-responsive common variant at chromosome band 7p21.1 confers tanning response and melanoma risk via regulation of the aryl hydrocarbon receptor, AHR. Am J Hum Genet 2021; 108:1611-1630. [PMID: 34343493 DOI: 10.1016/j.ajhg.2021.07.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022] Open
Abstract
Genome-wide association studies (GWASs) have identified a melanoma-associated locus on chromosome band 7p21.1 with rs117132860 as the lead SNP and a secondary independent signal marked by rs73069846. rs117132860 is also associated with tanning ability and cutaneous squamous cell carcinoma (cSCC). Because ultraviolet radiation (UVR) is a key environmental exposure for all three traits, we investigated the mechanisms by which this locus contributes to melanoma risk, focusing on cellular response to UVR. Fine-mapping of melanoma GWASs identified four independent sets of candidate causal variants. A GWAS region-focused Capture-C study of primary melanocytes identified physical interactions between two causal sets and the promoter of the aryl hydrocarbon receptor (AHR). Subsequent chromatin state annotation, eQTL, and luciferase assays identified rs117132860 as a functional variant and reinforced AHR as a likely causal gene. Because AHR plays critical roles in cellular response to dioxin and UVR, we explored links between this SNP and AHR expression after both 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) and ultraviolet B (UVB) exposure. Allele-specific AHR binding to rs117132860-G was enhanced following both, consistent with predicted weakened AHR binding to the risk/poor-tanning rs117132860-A allele, and allele-preferential AHR expression driven from the protective rs117132860-G allele was observed following UVB exposure. Small deletions surrounding rs117132860 introduced via CRISPR abrogates AHR binding, reduces melanocyte cell growth, and prolongs growth arrest following UVB exposure. These data suggest AHR is a melanoma susceptibility gene at the 7p21.1 risk locus and rs117132860 is a functional variant within a UVB-responsive element, leading to allelic AHR expression and altering melanocyte growth phenotypes upon exposure.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD 20892, USA.
| |
Collapse
|
7
|
Abstract
Variants in the Melanocortin 1 Receptor (MC1R) gene have been associated with an increased risk of melanoma, but the role in nevus count is unclear. We investigated if specific MC1R gene variants or the number of MC1R gene variants and phenotypical features were associated with nevus count. A total of 494 participants of the 'Leiden skin cancer study' were included and the MC1R gene coding sequence was analysed by single-strand conformation polymorphism analysis followed by sequencing of unknown variants. The association between MC1R gene variants and nevus count and the association between age, gender and phenotypical features and nevus count were studied using the Chi-square test. Study of nine frequently occurring MC1R gene variants in participants without skin cancer (n = 203) showed that the 'r' Val60Leu variant was significantly associated with high nevus count (>50 nevi) (P = 0.017). This association was very strong among women (P < 0.001), but not present among men. Having one or two MC1R variants in general did not show a significant difference in the nevus count. Hair colour, skin type, eye colour and age were not significantly associated with nevus count, whereas gender showed a significant association (P = 0.008), with the highest nevus counts in female. The Val60Leu variant of the MC1R gene could be a promising candidate as an independent predictor of high nevus count, particularly in women. This information about the genetic makeup could promote personalized follow-up strategies and might help to prevent skin cancer in the future.
Collapse
|
8
|
Podlipnik S, Potrony M, Puig S. Genetic markers for characterization and prediction of prognosis of melanoma subtypes: a 2021 update. Ital J Dermatol Venerol 2021; 156:322-330. [PMID: 33982545 DOI: 10.23736/s2784-8671.21.06957-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
In this article we examined the most important genetic markers involved in melanoma susceptibility, initiation and progression, and their impact on the prognosis of the disease. Current knowledge in melanoma genetics identifies distinct pathways to the development of different melanoma subtypes characterized by specific clinico-pathological features and partially known genetic markers, modulated by high, low or absence of cumulative sun damage. The most prevalent somatic mutations are related to the activation of the MAPK pathway, which are classified into four major subtypes: BRAF mutant, NRAS mutant, NF1 mutant and triple wild type. Moreover, germinal mutations are also involved in the characterization and predictions of prognosis in melanoma. Currently, CDKN2A is seen as the main high-risk gene involved in melanoma susceptibility being mutated in around 20% of melanoma-prone families. Other high-risk susceptibility genes described include CDK4, POT1, BAP1, TERT promoter, ACD, and TERF2IP. Melanoma is one of the most genetically predisposed among all cancers in humans, and ultraviolet light from the sun is the main environmental factor. This genetic predisposition is starting to be understood, impacting not only on the risk of developing melanoma but also on the risk of developing other types of cancer, as well as on the prognosis of the disease.
Collapse
Affiliation(s)
- Sebastian Podlipnik
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain.,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain
| | - Miriam Potrony
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain.,Department of Biochemistry and Molecular Genetics, Hospital of Barcelona, Barcelona, Spain
| | - Susana Puig
- Department of Dermatology, University of Barcelona, Hospital of Barcelona, Barcelona, Spain - .,Unit of Melanoma, Institut d'Investigacions Biomèdiques August Pi I Sunyer (IDIBAPS), Barcelona, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Barcelona, Spain
| |
Collapse
|
9
|
Gene Discovery Using Twins. Twin Res Hum Genet 2021; 23:90-93. [PMID: 32638676 DOI: 10.1017/thg.2020.38] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
One of Nick's key early achievements at QIMR was to establish a twin study on melanoma risk factors. The Brisbane Twin Nevus Study (BTNS) had an initial focus on nevus (mole) count in adolescents but, reflecting Nick's broad interests, expanded in scope enormously over the decades. In the skin cancer arena, BTNS was essential to genetic discoveries in melanoma, eye color and pigmentation. Later studies amassed data on thousands of phenotypes, ranging from molecular phenotypes such as gene expression to studies where gene mapping findings in adolescents turned out to have translational potential in late-onset diseases. Nick's twin data have formed the basis for an enormous range of discoveries, with Nick and his colleagues continuing to capitalize on these data.
Collapse
|
10
|
|
11
|
Pozzobon FC, Tell-Marti G, Calbet-Llopart N, Barreiro A, Espinosa N, Potrony M, Alejo B, Podlipnik S, Combalia M, Puig-Butillé JA, Carrera C, Malvehy J, Puig S. Influence of germline genetic variants on dermoscopic features of melanoma. Pigment Cell Melanoma Res 2021; 34:618-628. [PMID: 33342058 DOI: 10.1111/pcmr.12954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2020] [Revised: 12/02/2020] [Accepted: 12/11/2020] [Indexed: 01/12/2023]
Abstract
Nevus count is highly determined by inherited variants and has been associated with the origin of melanoma. De novo melanomas (DNMMs) are more prevalent in patients with a low nevus count and have distinctive dermoscopic features than nevus-associated melanomas. We evaluated the impact of nine single nucleotide polymorphisms (SNPs) of MTAP (rs10811629, rs2218220, rs7023329 and rs751173), PLA2G6 (rs132985 and rs2284063), IRF4 (rs12203592), and PAX3 (rs10180903 and rs7600206) genes associated with nevus count and melanoma susceptibility, and the MC1R variants on dermoscopic features of 371 melanomas from 310 patients. All MTAP variants associated with a low nevus count were associated with regression structures (peppering and mixed regression), blue-whitish veil, shiny white structures, and pigment network. SNPs of PLA2G6 (rs132985), PAX3 (rs7600206), and IRF4 (rs12203592) genes were also associated with either shiny white structures or mixed regression (all corrected p-values ≤ .06). Melanomas from red hair color MC1R variants carriers showed lower total dermoscopy score (p-value = .015) and less blotches than melanomas from non-carriers (p-value = .048). Our results provide evidence that germline variants protective for melanoma risk and/or associated with a low nevus count are associated with certain dermoscopic features, more characteristic of de novo and worse prognosis melanomas.
Collapse
Affiliation(s)
- Flavia Carolina Pozzobon
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain.,Universidad Nacional de Colombia, Bogotá, Colombia
| | - Gemma Tell-Marti
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Neus Calbet-Llopart
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Alicia Barreiro
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
| | - Natalia Espinosa
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
| | - Míriam Potrony
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Beatriz Alejo
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
| | - Sebastian Podlipnik
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
| | - Marc Combalia
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
| | - Joan Anton Puig-Butillé
- Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain.,Biochemical and Molecular Genetics Service, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain
| | - Cristina Carrera
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain
| | - Josep Malvehy
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain.,Medicine Department, Universitat de Barcelona, Barcelona, Spain
| | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clínic & IDIBAPS (Institut d'Investigacions Biomèdiques August Pi i Sunyer), Barcelona, Spain.,Biomedical Research Networking Center on Rare Diseases (CIBERER), ISCIII, Barcelona, Spain.,Medicine Department, Universitat de Barcelona, Barcelona, Spain
| |
Collapse
|
12
|
Lipid metabolic Reprogramming: Role in Melanoma Progression and Therapeutic Perspectives. Cancers (Basel) 2020; 12:cancers12113147. [PMID: 33121001 PMCID: PMC7692067 DOI: 10.3390/cancers12113147] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2020] [Revised: 10/21/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023] Open
Abstract
Simple Summary Melanoma is a devastating skin cancer characterized by an impressive metabolic plasticity. Melanoma cells are able to adapt to the tumor microenvironment by using a variety of fuels that contribute to tumor growth and progression. In this review, the authors summarize the contribution of the lipid metabolic network in melanoma plasticity and aggressiveness, with a particular attention to specific lipid classes such as glycerophospholipids, sphingolipids, sterols and eicosanoids. They also highlight the role of adipose tissue in tumor progression as well as the potential antitumor role of drugs targeting critical steps of lipid metabolic pathways in the context of melanoma. Abstract Metabolic reprogramming contributes to the pathogenesis and heterogeneity of melanoma. It is driven both by oncogenic events and the constraints imposed by a nutrient- and oxygen-scarce microenvironment. Among the most prominent metabolic reprogramming features is an increased rate of lipid synthesis. Lipids serve as a source of energy and form the structural foundation of all membranes, but have also emerged as mediators that not only impact classical oncogenic signaling pathways, but also contribute to melanoma progression. Various alterations in fatty acid metabolism have been reported and can contribute to melanoma cell aggressiveness. Elevated expression of the key lipogenic fatty acid synthase is associated with tumor cell invasion and poor prognosis. Fatty acid uptake from the surrounding microenvironment, fatty acid β-oxidation and storage also appear to play an essential role in tumor cell migration. The aim of this review is (i) to focus on the major alterations affecting lipid storage organelles and lipid metabolism. A particular attention has been paid to glycerophospholipids, sphingolipids, sterols and eicosanoids, (ii) to discuss how these metabolic dysregulations contribute to the phenotype plasticity of melanoma cells and/or melanoma aggressiveness, and (iii) to highlight therapeutic approaches targeting lipid metabolism that could be applicable for melanoma treatment.
Collapse
|
13
|
Fang S, Lu J, Zhou X, Wang Y, Ross MI, Gershenwald JE, Cormier JN, Wargo J, Sui D, Amos CI, Lee JE. Functional annotation of melanoma risk loci identifies novel susceptibility genes. Carcinogenesis 2020; 41:452-457. [PMID: 31630191 DOI: 10.1093/carcin/bgz173] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Revised: 09/23/2019] [Accepted: 10/15/2019] [Indexed: 12/27/2022] Open
Abstract
Genome-wide association study (GWAS)-identified single-nucleotide polymorphisms (SNPs) are tag SNPs located in both transcribed and non-coding regulatory DNA regions, rather than representing causal or functional variants for disease. To identify functional variants or genes for melanoma susceptibility, we used functional mapping and annotation (FUMA) to perform functional annotation of the summary statistics of 2541 significant melanoma risk SNPs (P < 5 × 10-8) identified by GWAS. The original GWAS melanoma study included 15 990 cases and 26 409 controls, representing the largest international meta-analysis of melanoma susceptibility. We prioritized 330 unique genes, including those in immune cytokine signaling pathways, from 19 loci through positional, expression quantitative trait locus, and chromatin interaction mapping. In comparison, only 38 melanoma-related genes were identified in the original meta-analysis. In addition to the well-known melanoma susceptibility genes confirmed in the meta-analysis (MC1R, CDKN2A, TERT, OCA2 and ARNT/SETDB1), we also identified additional novel genes using FUMA to map SNPs to genes. Through chromatin interaction mapping, we prioritized IFNA7, IFNA10, IFNA16, IFNA17, IFNA14, IFNA6, IFNA21, IFNA4, IFNE and IFNA5; these 10 most significant genes are all involved in immune system and cytokine signaling pathways. In the gene analysis, we identified 72 genes with a P < 2.5 × 10-6. The genes associated with melanoma risk were DEF8 (P = 1.09 × 10-57), DBNDD1 (P = 2.19 × 10-42), SPATA33 (P = 3.54 × 10-38) and MC1R (P = 1.04 × 10-36). In summary, this study identifies novel putative melanoma susceptibility genes and provides a guide for further experimental validation of functional variants and disease-related genes.
Collapse
Affiliation(s)
- Shenying Fang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jiachun Lu
- The Institute for Chemical Carcinogenesis, Collaborative Innovation Center for Environmental Toxicity, School of Public Health, Guangzhou Medical University, Guangzhou, China
| | - Xinke Zhou
- The Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yuling Wang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Merrick I Ross
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jeffrey E Gershenwald
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Janice N Cormier
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jennifer Wargo
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Dawen Sui
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | | | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
14
|
Choi J, Zhang T, Vu A, Ablain J, Makowski MM, Colli LM, Xu M, Hennessey RC, Yin J, Rothschild H, Gräwe C, Kovacs MA, Funderburk KM, Brossard M, Taylor J, Pasaniuc B, Chari R, Chanock SJ, Hoggart CJ, Demenais F, Barrett JH, Law MH, Iles MM, Yu K, Vermeulen M, Zon LI, Brown KM. Massively parallel reporter assays of melanoma risk variants identify MX2 as a gene promoting melanoma. Nat Commun 2020; 11:2718. [PMID: 32483191 PMCID: PMC7264232 DOI: 10.1038/s41467-020-16590-1] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2019] [Accepted: 05/12/2020] [Indexed: 02/07/2023] Open
Abstract
Genome-wide association studies (GWAS) have identified ~20 melanoma susceptibility loci, most of which are not functionally characterized. Here we report an approach integrating massively-parallel reporter assays (MPRA) with cell-type-specific epigenome and expression quantitative trait loci (eQTL) to identify susceptibility genes/variants from multiple GWAS loci. From 832 high-LD variants, we identify 39 candidate functional variants from 14 loci displaying allelic transcriptional activity, a subset of which corroborates four colocalizing melanocyte cis-eQTL genes. Among these, we further characterize the locus encompassing the HIV-1 restriction gene, MX2 (Chr21q22.3), and validate a functional intronic variant, rs398206. rs398206 mediates the binding of the transcription factor, YY1, to increase MX2 levels, consistent with the cis-eQTL of MX2 in primary human melanocytes. Melanocyte-specific expression of human MX2 in a zebrafish model demonstrates accelerated melanoma formation in a BRAFV600E background. Our integrative approach streamlines GWAS follow-up studies and highlights a pleiotropic function of MX2 in melanoma susceptibility. There are more than 20 known melanoma susceptibility genes. Here, using a massively parallel reporter assay, the authors identify risk-associated variants that alter gene transcription, and demonstrate that expression of one such gene, MX2, leads to the promotion of melanoma in a zebrafish model.
Collapse
Affiliation(s)
- Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Andrew Vu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Julien Ablain
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Matthew M Makowski
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 XZ, Nijmegen, The Netherlands
| | - Leandro M Colli
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Mai Xu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Rebecca C Hennessey
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Jinhu Yin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Harriet Rothschild
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Cathrin Gräwe
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 XZ, Nijmegen, The Netherlands
| | - Michael A Kovacs
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Karen M Funderburk
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Myriam Brossard
- Université de Paris, UMRS-1124, Institut National de la Santé et de la Recherche Médicale (INSERM), F-75006, Paris, France
| | - John Taylor
- Leeds Institute for Data Analytics, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Bogdan Pasaniuc
- Department of Human Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90024, USA
| | - Raj Chari
- Genome Modification Core, Frederick National Lab for Cancer Research, National Cancer Institute, Frederick, MD, 21701, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Clive J Hoggart
- Department of Medicine, Imperial College London, London, SW7 2BU, UK
| | - Florence Demenais
- Université de Paris, UMRS-1124, Institut National de la Santé et de la Recherche Médicale (INSERM), F-75006, Paris, France
| | - Jennifer H Barrett
- Leeds Institute for Data Analytics, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, QLD, 4006, Australia
| | - Mark M Iles
- Leeds Institute for Data Analytics, School of Medicine, University of Leeds, Leeds, LS2 9JT, UK
| | - Kai Yu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, 6525 XZ, Nijmegen, The Netherlands
| | - Leonard I Zon
- Stem Cell Program and Division of Hematology/Oncology, Boston Children's Hospital and Dana-Farber Cancer Institute, Boston, MA, 02115, USA
| | - Kevin M Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, 20892, USA.
| |
Collapse
|
15
|
Landi MT, Bishop DT, MacGregor S, Machiela MJ, Stratigos AJ, Ghiorzo P, Brossard M, Calista D, Choi J, Fargnoli MC, Zhang T, Rodolfo M, Trower AJ, Menin C, Martinez J, Hadjisavvas A, Song L, Stefanaki I, Scolyer R, Yang R, Goldstein AM, Potrony M, Kypreou KP, Pastorino L, Queirolo P, Pellegrini C, Cattaneo L, Zawistowski M, Gimenez-Xavier P, Rodriguez A, Elefanti L, Manoukian S, Rivoltini L, Smith BH, Loizidou MA, Del Regno L, Massi D, Mandala M, Khosrotehrani K, Akslen LA, Amos CI, Andresen PA, Avril MF, Azizi E, Soyer HP, Bataille V, Dalmasso B, Bowdler LM, Burdon KP, Chen WV, Codd V, Craig JE, Dębniak T, Falchi M, Fang S, Friedman E, Simi S, Galan P, Garcia-Casado Z, Gillanders EM, Gordon S, Green A, Gruis NA, Hansson J, Harland M, Harris J, Helsing P, Henders A, Hočevar M, Höiom V, Hunter D, Ingvar C, Kumar R, Lang J, Lathrop GM, Lee JE, Li X, Lubiński J, Mackie RM, Malt M, Malvehy J, McAloney K, Mohamdi H, Molven A, Moses EK, Neale RE, Novaković S, Nyholt DR, Olsson H, Orr N, Fritsche LG, Puig-Butille JA, Qureshi AA, Radford-Smith GL, Randerson-Moor J, Requena C, Rowe C, Samani NJ, Sanna M, Schadendorf D, et alLandi MT, Bishop DT, MacGregor S, Machiela MJ, Stratigos AJ, Ghiorzo P, Brossard M, Calista D, Choi J, Fargnoli MC, Zhang T, Rodolfo M, Trower AJ, Menin C, Martinez J, Hadjisavvas A, Song L, Stefanaki I, Scolyer R, Yang R, Goldstein AM, Potrony M, Kypreou KP, Pastorino L, Queirolo P, Pellegrini C, Cattaneo L, Zawistowski M, Gimenez-Xavier P, Rodriguez A, Elefanti L, Manoukian S, Rivoltini L, Smith BH, Loizidou MA, Del Regno L, Massi D, Mandala M, Khosrotehrani K, Akslen LA, Amos CI, Andresen PA, Avril MF, Azizi E, Soyer HP, Bataille V, Dalmasso B, Bowdler LM, Burdon KP, Chen WV, Codd V, Craig JE, Dębniak T, Falchi M, Fang S, Friedman E, Simi S, Galan P, Garcia-Casado Z, Gillanders EM, Gordon S, Green A, Gruis NA, Hansson J, Harland M, Harris J, Helsing P, Henders A, Hočevar M, Höiom V, Hunter D, Ingvar C, Kumar R, Lang J, Lathrop GM, Lee JE, Li X, Lubiński J, Mackie RM, Malt M, Malvehy J, McAloney K, Mohamdi H, Molven A, Moses EK, Neale RE, Novaković S, Nyholt DR, Olsson H, Orr N, Fritsche LG, Puig-Butille JA, Qureshi AA, Radford-Smith GL, Randerson-Moor J, Requena C, Rowe C, Samani NJ, Sanna M, Schadendorf D, Schulze HJ, Simms LA, Smithers M, Song F, Swerdlow AJ, van der Stoep N, Kukutsch NA, Visconti A, Wallace L, Ward SV, Wheeler L, Sturm RA, Hutchinson A, Jones K, Malasky M, Vogt A, Zhou W, Pooley KA, Elder DE, Han J, Hicks B, Hayward NK, Kanetsky PA, Brummett C, Montgomery GW, Olsen CM, Hayward C, Dunning AM, Martin NG, Evangelou E, Mann GJ, Long G, Pharoah PDP, Easton DF, Barrett JH, Cust AE, Abecasis G, Duffy DL, Whiteman DC, Gogas H, De Nicolo A, Tucker MA, Newton-Bishop JA, Peris K, Chanock SJ, Demenais F, Brown KM, Puig S, Nagore E, Shi J, Iles MM, Law MH. Genome-wide association meta-analyses combining multiple risk phenotypes provide insights into the genetic architecture of cutaneous melanoma susceptibility. Nat Genet 2020; 52:494-504. [PMID: 32341527 PMCID: PMC7255059 DOI: 10.1038/s41588-020-0611-8] [Show More Authors] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2019] [Accepted: 03/09/2020] [Indexed: 12/17/2022]
Abstract
Most genetic susceptibility to cutaneous melanoma remains to be discovered. Meta-analysis genome-wide association study (GWAS) of 36,760 cases of melanoma (67% newly genotyped) and 375,188 controls identified 54 significant (P < 5 × 10-8) loci with 68 independent single nucleotide polymorphisms. Analysis of risk estimates across geographical regions and host factors suggests the acral melanoma subtype is uniquely unrelated to pigmentation. Combining this meta-analysis with GWAS of nevus count and hair color, and transcriptome association approaches, uncovered 31 potential secondary loci for a total of 85 cutaneous melanoma susceptibility loci. These findings provide insights into cutaneous melanoma genetic architecture, reinforcing the importance of nevogenesis, pigmentation and telomere maintenance, together with identifying potential new pathways for cutaneous melanoma pathogenesis.
Collapse
Affiliation(s)
- Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA.
| | - D Timothy Bishop
- Leeds Institute of Medical Research at St James's, Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mitchell J Machiela
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander J Stratigos
- Department of Dermatology, Andreas Syggros Hospital, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Paola Ghiorzo
- Genetics of Rare Cancers, Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Myriam Brossard
- Genetic Epidemiology and Functional Genomics of Multifactorial Diseases Team, Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-1124, Université Paris Descartes, Paris, France
| | - Donato Calista
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Jiyeon Choi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Maria Concetta Fargnoli
- Department of Dermatology & Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Monica Rodolfo
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Adam J Trower
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK
| | - Chiara Menin
- Immunology and Molecular Oncology Unit, Venito Institute of Oncology IOV-IRCCS, Padua, Italy
| | | | - Andreas Hadjisavvas
- Department of EM/Molecular Pathology & The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Lei Song
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Irene Stefanaki
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Richard Scolyer
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal Prince Alfred Hospital, Sydney, New South Wales, Australia
- Central Clinical School, The University of Sydney, Sydney, New South Wales, Australia
- New South Wales Health Pathology, Sydney, New South Wales, Australia
| | - Rose Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Miriam Potrony
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, CIBERER, Barcelona, Spain
| | - Katerina P Kypreou
- Department of Dermatology, University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Lorenza Pastorino
- Genetics of Rare Cancers, Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Paola Queirolo
- Medical Oncology Unit, IRCCS Ospedale Policlinico San Martino, Genoa, Italy
| | - Cristina Pellegrini
- Department of Dermatology & Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Laura Cattaneo
- Pathology Unit, Azienda Socio-Sanitaria Territoriale Papa Giovanni XXIII, Bergamo, Italy
| | - Matthew Zawistowski
- Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
- Center for Statistical Genetics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Pol Gimenez-Xavier
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, CIBERER, Barcelona, Spain
| | - Arantxa Rodriguez
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Lisa Elefanti
- Immunology and Molecular Oncology Unit, Venito Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Siranoush Manoukian
- Unit of Medical Genetics, Department of Medical Oncology and Hematology, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Licia Rivoltini
- Unit of Immunotherapy of Human Tumors, Department of Research, Fondazione IRCCS Istituto Nazionale dei Tumori di Milano, Milan, Italy
| | - Blair H Smith
- Division of Population Health and Genomics, Ninewells Hospital and Medical School, University of Dundee, Dundee, UK
| | - Maria A Loizidou
- Department of EM/Molecular Pathology & The Cyprus School of Molecular Medicine, The Cyprus Institute of Neurology and Genetics, Nicosia, Cyprus
| | - Laura Del Regno
- Institute of Dermatology, Catholic University, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Daniela Massi
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Mario Mandala
- Department of Oncology, Giovanni XXIII Hospital, Bergamo, Italy
| | - Kiarash Khosrotehrani
- UQ Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
| | - Lars A Akslen
- Centre for Cancer Biomarkers CCBIO, Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
| | - Christopher I Amos
- Department of Community and Family Medicine, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Per A Andresen
- Department of Pathology, Molecular Pathology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Marie-Françoise Avril
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin, Service de Dermatologie, Université Paris Descartes, Paris, France
| | - Esther Azizi
- Department of Dermatology, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv, Israel
- Oncogenetics Unit, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - H Peter Soyer
- Department of Dermatology, Princess Alexandra Hospital, Brisbane, Queensland, Australia
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Veronique Bataille
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Dermatology, West Herts NHS Trust, Herts, UK
| | - Bruna Dalmasso
- Genetics of Rare Cancers, Ospedale Policlinico San Martino, Genoa, Italy
- Department of Internal Medicine and Medical Specialties (DIMI), University of Genoa, Genoa, Italy
| | - Lisa M Bowdler
- Sample Processing, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Kathryn P Burdon
- Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Wei V Chen
- Department of Genetics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Veryan Codd
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Jamie E Craig
- Department of Ophthalmology, Flinders University, Adelaide, South Australia, Australia
| | - Tadeusz Dębniak
- Department of Genetics and Pathology, International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Dermatology, West Herts NHS Trust, Herts, UK
| | - Shenying Fang
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Eitan Friedman
- Oncogenetics Unit, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Sarah Simi
- Section of Anatomic Pathology, Department of Health Sciences, University of Florence, Florence, Italy
| | - Pilar Galan
- Université Paris 13, Equipe de Recherche en Epidémiologie Nutritionnelle (EREN), Centre de Recherche en Epidémiologie et Statistiques, Institut National de la Santé et de la Recherche Médicale (INSERM U1153), Institut National de la Recherche Agronomique (INRA U1125), Conservatoire National des Arts et Métiers, Communauté d'Université Sorbonne Paris Cité, Bobigny, France
| | - Zaida Garcia-Casado
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Elizabeth M Gillanders
- Inherited Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Baltimore, MD, USA
| | - Scott Gordon
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Adele Green
- Cancer and Population Studies, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- CRUK Manchester Institute, Institute of Inflammation and Repair, University of Manchester, Manchester, UK
| | - Nelleke A Gruis
- Department of Dermatology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - Mark Harland
- Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK
| | - Jessica Harris
- Translational Research Institute, Institute of Health and Biomedical Innovation, Princess Alexandra Hospital, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Per Helsing
- Department of Dermatology, Oslo University Hospital, Rikshospitalet, Oslo, Norway
| | - Anjali Henders
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Marko Hočevar
- Department of Surgical Oncology, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Karolinska University Hospital, Stockholm, Sweden
| | - David Hunter
- Nuffield Department of Population Health, University of Oxford, Oxford, UK
- Program in Genetic Epidemiology and Statistical Genetics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Christian Ingvar
- Department of Surgery, Clinical Sciences, Lund University, Lund, Sweden
| | - Rajiv Kumar
- Division of Molecular Genetic Epidemiology, German Cancer Research Center, Heidelberg, Germany
| | - Julie Lang
- Department of Medical Genetics, University of Glasgow, Glasgow, UK
| | - G Mark Lathrop
- McGill University and Genome Quebec Innovation Centre, Montreal, Canada
| | - Jeffrey E Lee
- Department of Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Xin Li
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Jan Lubiński
- International Hereditary Cancer Center, Pomeranian Medical University, Szczecin, Poland
| | - Rona M Mackie
- Department of Medical Genetics, University of Glasgow, Glasgow, UK
- Department of Public Health, University of Glasgow, Glasgow, UK
| | - Maryrose Malt
- Cancer and Population Studies, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Josep Malvehy
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, CIBERER, Barcelona, Spain
| | - Kerrie McAloney
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Hamida Mohamdi
- Genetic Epidemiology and Functional Genomics of Multifactorial Diseases Team, Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-1124, Université Paris Descartes, Paris, France
| | - Anders Molven
- Department of Pathology, Haukeland University Hospital, Bergen, Norway
- Gade Laboratory for Pathology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Eric K Moses
- Centre for Genetic Origins of Health and Disease, Faculty of Medicine, Dentistry and Health Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Rachel E Neale
- Cancer Aetiology & Prevention, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Srdjan Novaković
- Department of Molecular Diagnostics, Institute of Oncology Ljubljana, Ljubljana, Slovenia
| | - Dale R Nyholt
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- School of Biomedical Sciences and Institute of Health and Biomedical Innovation, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Håkan Olsson
- Department of Oncology/Pathology, Clinical Sciences, Lund University, Lund, Sweden
- Department of Cancer Epidemiology, Clinical Sciences, Lund University, Lund, Sweden
| | - Nicholas Orr
- Breakthrough Breast Cancer Research Centre, The Institute of Cancer Research, London, UK
| | - Lars G Fritsche
- Center for Statistical Genetics, Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Joan Anton Puig-Butille
- Biochemistry and Molecular Genetics Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona,CIBERER, Barcelona, Spain
| | - Abrar A Qureshi
- Department of Dermatology, The Warren Alpert Medical School of Brown University, Providence, RI, USA
| | - Graham L Radford-Smith
- Inflammatory Bowel Diseases, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
- Department of Gastroenterology and Hepatology, Royal Brisbane & Women's Hospital, Brisbane, Queensland, Australia
- University of Queensland School of Medicine, Herston Campus, Brisbane, Queensland, Australia
| | | | - Celia Requena
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Casey Rowe
- UQ Diamantina Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Nilesh J Samani
- Department of Cardiovascular Sciences, University of Leicester, Leicester, UK
- NIHR Leicester Biomedical Research Centre, Glenfield Hospital, Leicester, UK
| | - Marianna Sanna
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Dermatology, West Herts NHS Trust, Herts, UK
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, Essen, Germany
- German Consortium Translational Cancer Research (DKTK), Heidelberg, Germany
| | - Hans-Joachim Schulze
- Department of Dermatology, Fachklinik Hornheide, Institute for Tumors of the Skin, University of Münster, Münster, Germany
| | - Lisa A Simms
- Inflammatory Bowel Diseases, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mark Smithers
- Queensland Melanoma Project, Princess Alexandra Hospital, The University of Queensland, St Lucia, Queensland, Australia
- Mater Research Institute, The University of Queensland, St Lucia, Queensland, Australia
| | - Fengju Song
- Departments of Epidemiology and Biostatistics, Key Laboratory of Cancer Prevention and Therapy, Tianjin, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, P. R. China
| | - Anthony J Swerdlow
- Division of Genetics and Epidemiology, The Institute of Cancer Research, London, UK
- Division of Breast Cancer Research, The Institute of Cancer Research, London, UK
| | - Nienke van der Stoep
- Department of Clinical Genetics, Center of Human and Clinical Genetics, Leiden University Medical Center, Leiden, the Netherlands
| | - Nicole A Kukutsch
- Department of Dermatology, Leiden University Medical Centre, Leiden, the Netherlands
| | - Alessia Visconti
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
- Department of Dermatology, West Herts NHS Trust, Herts, UK
| | - Leanne Wallace
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Sarah V Ward
- Centre for Genetic Origins of Health and Disease, School of Biomedical Sciences, The University of Western Australia, Perth, Western Australia, Australia
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Lawrie Wheeler
- Translational Research Institute, Institute of Health and Biomedical Innovation, Princess Alexandra Hospital, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Richard A Sturm
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
| | - Amy Hutchinson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Genome Research Laboratory, Leidos Biomedical Research, Bethesda, MD, USA
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Genome Research Laboratory, Leidos Biomedical Research, Bethesda, MD, USA
| | - Michael Malasky
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Genome Research Laboratory, Leidos Biomedical Research, Bethesda, MD, USA
| | - Aurelie Vogt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Genome Research Laboratory, Leidos Biomedical Research, Bethesda, MD, USA
| | - Weiyin Zhou
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Genome Research Laboratory, Leidos Biomedical Research, Bethesda, MD, USA
| | - Karen A Pooley
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - David E Elder
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Cancer Genome Research Laboratory, Leidos Biomedical Research, Bethesda, MD, USA
| | - Nicholas K Hayward
- Oncogenomics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Chad Brummett
- Department of Anesthesiology, University of Michigan, Ann Arbor, MI, USA
| | - Grant W Montgomery
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Catherine M Olsen
- Cancer Control Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh, UK
| | - Alison M Dunning
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Nicholas G Martin
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Graham J Mann
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Centre for Cancer Research, Westmead Institute for Medical Research, Sydney, Australia
- John Curtin School of Medical Research, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Georgina Long
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Royal North Shore Hospital, Sydney, Australia
| | - Paul D P Pharoah
- Department of Oncology, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | - Douglas F Easton
- Department of Public Health and Primary Care, Centre for Cancer Genetic Epidemiology, University of Cambridge, Cambridge, UK
| | | | - Anne E Cust
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
- Cancer Epidemiology and Prevention Research, Sydney School of Public Health, Sydney, Australia
| | - Goncalo Abecasis
- Department of Biostatistics, University of Michigan, Ann Arbor, MI, USA
| | - David L Duffy
- Dermatology Research Centre, The University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Queensland, Australia
- Genetic Epidemiology, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - David C Whiteman
- Cancer Control Group, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Helen Gogas
- First Department of Internal Medicine, Laikon General Hospital Greece, National and Kapodistrian University of Athens, Athens, Greece
| | - Arcangela De Nicolo
- Cancer Genomics Program, Veneto Institute of Oncology IOV-IRCCS, Padua, Italy
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Ketty Peris
- Institute of Dermatology, Catholic University, Rome, Italy
- Fondazione Policlinico Universitario A. Gemelli, IRCCS, Rome, Italy
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Florence Demenais
- Genetic Epidemiology and Functional Genomics of Multifactorial Diseases Team, Institut National de la Santé et de la Recherche Médicale (INSERM), UMRS-1124, Université Paris Descartes, Paris, France
| | - Kevin M Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Susana Puig
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, CIBERER, Barcelona, Spain
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Mark M Iles
- Leeds Institute for Data Analytics, University of Leeds, Leeds, UK.
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia.
| |
Collapse
|
16
|
Dalmasso B, Ghiorzo P. Evolution of approaches to identify melanoma missing heritability. Expert Rev Mol Diagn 2020; 20:523-531. [PMID: 32124637 DOI: 10.1080/14737159.2020.1738221] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Accepted: 03/02/2020] [Indexed: 02/08/2023]
Abstract
Introduction: Around 10% of melanoma patients have a positive family history of melanoma and/or related cancers. Although a germline pathogenic variant in a high-risk gene can be identified in up to 40% of these patients, the remaining part of melanoma heritability remains largely unexplained.Areas covered: The aim of this review is to provide an overview of the impact that new technologies and new research approaches had and are having on finding more efficient ways to unravel the missing heritability in melanoma.Expert opinion: High-throughput sequencing technologies have been crucial in increasing the number of genes/loci that might be implicated in melanoma predisposition. However, results from these approaches may have been inferior to the expectations, due to an increase in quantitative information which hasn't been followed at the same speed by an improvement of the methods to correctly interpret these data. Optimal approaches for improving our knowledge on melanoma heritability are currently based on segregation analysis coupled with functional assessment of candidate genes. An improvement of computational methods to infer genotype-phenotype correlations could help address the issue of missing heritability.
Collapse
Affiliation(s)
- Bruna Dalmasso
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa, Genova, Italy
- IRCCS Ospedale Policlinico San Martino, Genetics of Rare Cancers, Genoa, Italy
| |
Collapse
|
17
|
Cust A, Drummond M, Bishop D, Azizi L, Schmid H, Jenkins M, Hopper J, Armstrong B, Aitken J, Kefford R, Giles G, Demenais F, Goldstein A, Barrett J, Kanetsky P, Elder D, Mann G, Newton‐Bishop J. Associations of pigmentary and naevus phenotype with melanoma risk in two populations with comparable ancestry but contrasting levels of ambient sun exposure. J Eur Acad Dermatol Venereol 2019; 33:1874-1885. [PMID: 31087403 PMCID: PMC6800761 DOI: 10.1111/jdv.15680] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2018] [Accepted: 05/03/2019] [Indexed: 11/29/2022]
Abstract
BACKGROUND People at high risk of developing melanoma are usually identified by pigmentary and naevus phenotypes. OBJECTIVE We examined whether associations of these phenotypes with melanoma risk differed by ambient sun exposure or participant characteristics in two population-based, case-control studies with comparable ancestry but different ambient sun exposure. METHODS Data were analysed from 616 cases and 496 controls from the Australian Melanoma Family Study and 2012 cases and 504 controls from the Leeds (UK) case-control study. Questionnaire, interview and dermatological skin examination data were collected using the same measurement protocols. Relative risks were estimated as odds ratios using unconditional logistic regression, adjusted for potential confounders. RESULTS Hair and skin colour were the strongest pigmentary phenotype risk factors. All associations of pigmentary phenotype with melanoma risk were similar across countries. The median number of clinically assessed naevi was approximately three times higher in Australia than Leeds, but the relative risks for melanoma associated with each additional common or dysplastic naevus were higher for Leeds than Australia, especially for naevi on the upper and lower limbs. Higher naevus counts on the head and neck were associated with a stronger relative risk for melanoma for women than men. The two countries had similar relative risks for melanoma based on self-reported naevus density categories, but personal perceptions of naevus number differed by country. There was no consistent evidence of interactions between phenotypes on risk. CONCLUSIONS Classifying people at high risk of melanoma based on their number of naevi should ideally take into account their country of residence, type of counts (clinical or self-reported), body site on which the naevus counts are measured and sex. The presence of naevi may be a stronger indicator of a genetic predisposition in the UK than in Australia based on less opportunity for sun exposure to influence naevus development.
Collapse
Affiliation(s)
- A.E. Cust
- Cancer Epidemiology and Prevention ResearchSydney School of Public HealthThe University of SydneySydneyAustralia
- Melanoma Institute AustraliaThe University of SydneySydneyAustralia
| | - M. Drummond
- Cancer Epidemiology and Prevention ResearchSydney School of Public HealthThe University of SydneySydneyAustralia
- Melanoma Institute AustraliaThe University of SydneySydneyAustralia
| | - D.T. Bishop
- Section of Epidemiology and BiostatisticsLeeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - L. Azizi
- School of Mathematics and StatisticsThe University of SydneySydneyAustralia
| | - H. Schmid
- Centre for Cancer ResearchWestmead Institute for Medical ResearchThe University of SydneySydneyAustralia
| | - M.A. Jenkins
- Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global HealthThe University of MelbourneMelbourneAustralia
| | - J.L. Hopper
- Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global HealthThe University of MelbourneMelbourneAustralia
| | - B.K. Armstrong
- Cancer Epidemiology and Prevention ResearchSydney School of Public HealthThe University of SydneySydneyAustralia
| | - J.F. Aitken
- Viertel Centre for Research in Cancer ControlCancer Council QueenslandBrisbaneAustralia
| | - R.F. Kefford
- Melanoma Institute AustraliaThe University of SydneySydneyAustralia
- Macquarie University Health Sciences CentreMacquarie UniversitySydneyAustralia
| | - G.G. Giles
- Centre for Epidemiology and BiostatisticsMelbourne School of Population and Global HealthThe University of MelbourneMelbourneAustralia
- Cancer Epidemiology CentreCancer Council VictoriaMelbourneAustralia
| | - F. Demenais
- Genetic Variation and Human Diseases UnitUMR‐946INSERMUniversité Paris DiderotUniversité Sorbonne Paris CitéParisFrance
| | - A.M. Goldstein
- Human Genetics ProgramDivision of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMDUSA
| | - J.H. Barrett
- Section of Epidemiology and BiostatisticsLeeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| | - P.A. Kanetsky
- Cancer Epidemiology ProgramMoffitt Cancer CenterTampaFLUSA
| | - D.E. Elder
- Department of Pathology and Laboratory MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - G.J. Mann
- Melanoma Institute AustraliaThe University of SydneySydneyAustralia
- Centre for Cancer ResearchWestmead Institute for Medical ResearchThe University of SydneySydneyAustralia
| | - J.A. Newton‐Bishop
- Section of Epidemiology and BiostatisticsLeeds Institute of Cancer and PathologyUniversity of LeedsLeedsUK
| |
Collapse
|
18
|
Inherited Melanoma Risk Variants Associated with Histopathologically Amelanotic Melanoma. J Invest Dermatol 2019; 140:918-922.e7. [PMID: 31568773 DOI: 10.1016/j.jid.2019.09.006] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 09/05/2019] [Accepted: 09/09/2019] [Indexed: 01/26/2023]
|
19
|
Visconti A, Ribero S, Sanna M, Spector TD, Bataille V, Falchi M. Body site-specific genetic effects influence naevus count distribution in women. Pigment Cell Melanoma Res 2019; 33:326-333. [PMID: 31403758 DOI: 10.1111/pcmr.12820] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 02/05/2023]
Abstract
Body site is highly relevant for melanoma: it affects prognosis and varies according to the patient's sex. The distribution of naevi, a major risk factor for melanoma, at different body sites also varies according to sex in childhood. Using naevus counts at different body sites in 492 unrelated adults from both sexes, we observed that women have an increased number of naevi on the lower limbs compared to men (p = 8.5 × 10-5 ), showing that a high naevus count on this site persists from childhood throughout life. Then, using data from 3,232 twins, we observed, in women, the lowest naevus count heritability on the trunk (26%), and the highest on the lower limbs (69%). Finally, we showed that, in 2,864 women, six genomic loci previously associated with both naevus count and melanoma risk (IRF4, DOCK8, MTAP, 9q31.2, KITLG and PLA2G6) have an effect on naevus count that is body site-specific, but whose effect sizes are predominantly stronger on the lower limbs. Sex-specific genetic influence on naevus count at different sites may explain differences in site-specific melanoma incidence as well as prognosis between sexes.
Collapse
Affiliation(s)
- Alessia Visconti
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Simone Ribero
- Department of Medical Sciences, Section of Dermatology, University of Turin, Turin, Italy
| | - Marianna Sanna
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Tim D Spector
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| | - Veronique Bataille
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK.,Department of Dermatology, West Herts NHS Trust, Herts, UK
| | - Mario Falchi
- Department of Twin Research & Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
20
|
Rayner JE, McMeniman EK, Duffy DL, De'Ambrosis B, Smithers BM, Jagirdar K, Lee KJ, Soyer HP, Sturm RA. Phenotypic and genotypic analysis of amelanotic and hypomelanotic melanoma patients. J Eur Acad Dermatol Venereol 2019; 33:1076-1083. [PMID: 30680790 DOI: 10.1111/jdv.15446] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2018] [Accepted: 12/19/2018] [Indexed: 01/18/2023]
Abstract
BACKGROUND Amelanotic/hypomelanotic melanoma is associated with poorer outcomes due to a more advanced disease stage at diagnosis. OBJECTIVE To determine phenotypic risks and genotypic associations with amelanotic/hypomelanotic melanoma to develop a clinical and genetic profile that could assist in identifying high-risk individuals. METHODS The Brisbane Naevus Morphology Study conducted from 2009 to 2016 has recruited a core of 1254 participants. Participants were drawn from a combination of volunteers from dermatology outpatient clinics, private dermatology clinics, the Brisbane Longitudinal Twin Study and QSkin study. Case participants had a personal history of melanoma and control participants no personal history of melanoma. We specifically examined seven known candidate pigmentation and melanoma genes and pigmentary phenotypic characteristics in participants with amelanotic/hypomelanotic melanoma compared to pigmented melanomas. This assayed single nucleotide polymorphisms in MC1R, TYR, HERC/OCA2, IRF4, MTAP, PLA2G6 and MITF. RESULTS Forty-seven participants had at least one amelanotic/hypomelanotic melanoma, and 389 had pigmented melanomas, with amelanotic/hypomelanotic melanoma patients significantly older than pigmented melanoma participants (63.3 ± 13.0 vs. 54.6 ± 15.3 years; P < 0.001). Amelanotic/hypomelanotic melanoma patients were more likely than pigmented melanoma patients to have red hair (34% vs. 15%; P = 0.01), severe hand freckling (13% vs. 5%; P = 0.01) and propensity to sunburn (63% vs. 44%; P = 0.01). MC1R R/R genotype was much more frequent in our amelanotic/hypomelanotic melanoma population (31.1% vs. 11%; P < 0.001; OR 26.4 vs. 5.9; control 1.0). Amelanotic/hypomelanotic melanoma was associated with TYR rs1126809*A/A [OR (CI 95%) 2.7 (1.1-6.8) vs. 1.2 (0.8-1.9)] and PLA2G6 rs11570734*A/A [OR (CI 95%) 3.7 (1.0-13.6) vs. 1.3 (0.9-2.0)]. The MTAP melanoma risk SNP genotype, associated with darker pigmentation, (rs4636294*A/A) was less common in amelanotic/hypomelanotic melanoma patients [OR (CI 95%) 0.8 (0.3-2.1) vs. 2.0 (1.3-3.1)]. CONCLUSIONS Knowledge of phenotypic and genotypic associations of amelanotic/hypomelanotic melanoma can help predict risks and associations of this difficult to diagnose melanoma, which may ultimately assist clinical management and patient skin self-examination.
Collapse
Affiliation(s)
- J E Rayner
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - E K McMeniman
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
- Dermatology Department, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - D L Duffy
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
- QIMR Berghofer Medical Research Institute, Brisbane, Qld, Australia
| | - B De'Ambrosis
- Dermatology Department, Princess Alexandra Hospital, Brisbane, Qld, Australia
- Faculty of Medicine, The University of Queensland, Brisbane, Qld, Australia
- South East Dermatology, Annerley Square, Brisbane, Qld, Australia
| | - B M Smithers
- Queensland Melanoma Project, School of Medicine, The University of Queensland, Brisbane, Qld, Australia
| | - K Jagirdar
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - K J Lee
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| | - H P Soyer
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
- Dermatology Department, Princess Alexandra Hospital, Brisbane, Qld, Australia
| | - R A Sturm
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Qld, Australia
| |
Collapse
|
21
|
Shi Z, Yu H, Wu Y, Lin X, Bao Q, Jia H, Perschon C, Duggan D, Helfand BT, Zheng SL, Xu J. Systematic evaluation of cancer-specific genetic risk score for 11 types of cancer in The Cancer Genome Atlas and Electronic Medical Records and Genomics cohorts. Cancer Med 2019; 8:3196-3205. [PMID: 30968590 PMCID: PMC6558466 DOI: 10.1002/cam4.2143] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2019] [Revised: 03/01/2019] [Accepted: 03/18/2019] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Genetic risk score (GRS) is an odds ratio (OR)-weighted and population-standardized method for measuring cumulative effect of multiple risk-associated single nucleotide polymorphisms (SNPs). We hypothesize that GRS is a valid tool for risk assessment of most common cancers. METHODS Utilizing genotype and phenotype data from The Cancer Genome Atlas (TCGA) and Electronic Medical Records and Genomics (eMERGE), we tested 11 cancer-specific GRSs (bladder, breast, colorectal, glioma, lung, melanoma, ovarian, pancreatic, prostate, renal, and thyroid cancer) for association with the respective cancer type. Cancer-specific GRSs were calculated, for the first time in these cohorts, based on previously published risk-associated SNPs using the Caucasian subjects in these two cohorts. RESULTS Mean cancer-specific GRS in the population controls of eMERGE approximated the expected value of 1.00 (between 0.98 and 1.02) for all 11 types of cancer. Mean cancer-specific GRS was consistently higher in respective cancer patients than controls for all 11 types of cancer (P < 0.05). When subjects were categorized into low-, average-, and high-risk groups based on cancer-specific GRS (<0.5, 0.5-1.5, and >1.5, respectively), significant dose-response associations of higher cancer-specific GRS with higher OR of respective type of cancer were found for nine types of cancer (P-trend < 0.05). More than 64% subjects in the population controls of eMERGE can be classified as high risk for at least one type of these cancers. CONCLUSION Validity of GRS for predicting cancer risk is demonstrated for most types of cancer. If confirmed in larger studies, cancer-specific GRS may have the potential for developing personalized cancer screening strategy.
Collapse
Affiliation(s)
- Zhuqing Shi
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois.,State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Hongjie Yu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Yishuo Wu
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoling Lin
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Quanwa Bao
- State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China
| | - Haifei Jia
- Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Chelsea Perschon
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - David Duggan
- Translational Genomics Research Institute, Phoenix, Arizona
| | - Brian T Helfand
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Siqun L Zheng
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois
| | - Jianfeng Xu
- Program for Personalized Cancer Care, NorthShore University HealthSystem, Evanston, Illinois.,State Key Laboratory of Genetic Engineering, School of Life Science, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
22
|
Melanoma susceptibility variant rs869330 in the MTAP gene is associated with melanoma outcome. Melanoma Res 2019; 29:590-595. [PMID: 30681428 DOI: 10.1097/cmr.0000000000000578] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
The rising incidence of cutaneous melanoma (CM), an aggressive skin cancer, emphasizes the need for novel biomarkers to guide personalized care and better predict outcome. Genetic factors including germline risk variants are promising candidates for this aim. We explored the association between germline risk variants and melanoma outcome in a large genetically homogenous Belgian melanoma population, focusing on single nucleotide polymorphisms which generated the highest association with melanoma susceptibility. Between 2004 and 2014, blood samples of 1088 patients with histologically confirmed CM were collected and genotyped for nine variants. Cox proportional hazard models were used to assess the association between each single nucleotide polymorphism and relapse-free survival and overall survival, adjusted by age, sex, melanoma stage, site, and subtype. We identified significant associations for rs869330 (in the methylthioadenosine phosphorylase - MTAP gene) with overall survival (hazard ratio = 0.760, P = 0.048, 95% confidence interval: 0.580-0.998) and relapse-free survival (hazard ratio = 0.800, P = 0.020, 95% confidence interval: 0.650-0.970). This exploratory study is the first to show a significant association between the rs869330 variant (in the MTAP gene) and outcome in a large CM population.
Collapse
|
23
|
Gu F, Chen TH, Pfeiffer RM, Fargnoli MC, Calista D, Ghiorzo P, Peris K, Puig S, Menin C, De Nicolo A, Rodolfo M, Pellegrini C, Pastorino L, Evangelou E, Zhang T, Hua X, DellaValle CT, Timothy Bishop D, MacGregor S, Iles MI, Law MH, Cust A, Brown KM, Stratigos AJ, Nagore E, Chanock S, Shi J, Consortium MMA, Consortium M, Landi MT. Combining common genetic variants and non-genetic risk factors to predict risk of cutaneous melanoma. Hum Mol Genet 2018; 27:4145-4156. [PMID: 30060076 PMCID: PMC6240742 DOI: 10.1093/hmg/ddy282] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2018] [Revised: 06/14/2018] [Accepted: 07/24/2018] [Indexed: 02/04/2023] Open
Abstract
Melanoma heritability is among the highest for cancer and single nucleotide polymorphisms (SNPs) contribute to it. To date, only SNPs that reached statistical significance in genome-wide association studies or few candidate SNPs have been included in melanoma risk prediction models. We compared four approaches for building polygenic risk scores (PRS) using 12 874 melanoma cases and 23 203 controls from Melanoma Meta-Analysis Consortium as a training set, and newly genotyped 3102 cases and 2301 controls from the MelaNostrum consortium for validation. We estimated adjusted odds ratios (ORs) for melanoma risk using traditional melanoma risk factors and the PRS with the largest area under the receiver operator characteristics curve (AUC). We estimated absolute risks combining the PRS and other risk factors, with age- and sex-specific melanoma incidence and competing mortality rates from Italy as an example. The best PRS, including 204 SNPs (AUC = 64.4%; 95% confidence interval (CI) = 63-65.8%), developed using winner's curse estimate corrections, had a per-quintile OR = 1.35 (95% CI = 1.30-1.41), corresponding to a 3.33-fold increase comparing the 5th to the 1st PRS quintile. The AUC improvement by adding the PRS was up to 7%, depending on adjusted factors and country. The 20-year absolute risk estimates based on the PRS, nevus count and pigmentation characteristics for a 60-year-old Italian man ranged from 0.5 to 11.8% (relative risk = 26.34), indicating good separation.
Collapse
Affiliation(s)
- Fangyi Gu
- Department of Cancer Prevention and Control, Roswell Park Comprehensive Cancer Center, Buffalo, NY, USA
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Ting-Huei Chen
- Department of Mathematics and Statistics, Laval University, Quebec, Canada
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | - Donato Calista
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Paola Ghiorzo
- Department of Internal Medicine and Medical Specialties, University of Genoa and Genetics of Rare Cancers, Ospedale Policlinico San Martino, Genoa, Italy
| | - Ketty Peris
- Institute of Dermatology, Catholic University, Rome, Italy
| | - Susana Puig
- Dermatology Department, Melanoma Unit, Hospital Clínic de Barcelona, IDIBAPS, Universitat de Barcelona, Barcelona, Spain and Centro de Investigación Biomédica en Red en Enfermedades Raras (CIBERER), Valencia, Spain
| | - Chiara Menin
- Department of Immunology and Molecular Oncology, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Arcangela De Nicolo
- Cancer Genomics Program, Veneto Institute of Oncology IOV–IRCCS, Padua, Italy
| | - Monica Rodolfo
- Department of Experimental Oncology and Molecular Medicine, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | | | - Lorenza Pastorino
- Department of Internal Medicine and Medical Specialties, University of Genoa and Genetics of Rare Cancers, Ospedale Policlinico San Martino, Genoa, Italy
| | - Evangelos Evangelou
- Department of Hygiene and Epidemiology, University of Ioannina Medical School, Ioannina, Greece
- Department of Epidemiology and Biostatistics, Imperial College London, London, UK
| | - Tongwu Zhang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xing Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Curt T DellaValle
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Mark I Iles
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, UK
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, Australia
| | - Anne Cust
- Sydney School of Public Health, and Melanoma Institute Australia, The University of Sydney, Sydney, Australia
| | - Kevin M Brown
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Alexander J Stratigos
- 1 Department of Dermatology–Venereology, National and Kapodistrian University of Athens School of Medicine, Andreas Sygros Hospital, Athens, Greece
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncología, València, Spain
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jianxin Shi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | | | | | - Maria Teresa Landi
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
24
|
Duffy DL, Zhu G, Li X, Sanna M, Iles MM, Jacobs LC, Evans DM, Yazar S, Beesley J, Law MH, Kraft P, Visconti A, Taylor JC, Liu F, Wright MJ, Henders AK, Bowdler L, Glass D, Ikram MA, Uitterlinden AG, Madden PA, Heath AC, Nelson EC, Green AC, Chanock S, Barrett JH, Brown MA, Hayward NK, MacGregor S, Sturm RA, Hewitt AW, Kayser M, Hunter DJ, Newton Bishop JA, Spector TD, Montgomery GW, Mackey DA, Smith GD, Nijsten TE, Bishop DT, Bataille V, Falchi M, Han J, Martin NG. Novel pleiotropic risk loci for melanoma and nevus density implicate multiple biological pathways. Nat Commun 2018; 9:4774. [PMID: 30429480 PMCID: PMC6235897 DOI: 10.1038/s41467-018-06649-5] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/13/2018] [Indexed: 11/09/2022] Open
Abstract
The total number of acquired melanocytic nevi on the skin is strongly correlated with melanoma risk. Here we report a meta-analysis of 11 nevus GWAS from Australia, Netherlands, UK, and USA comprising 52,506 individuals. We confirm known loci including MTAP, PLA2G6, and IRF4, and detect novel SNPs in KITLG and a region of 9q32. In a bivariate analysis combining the nevus results with a recent melanoma GWAS meta-analysis (12,874 cases, 23,203 controls), SNPs near GPRC5A, CYP1B1, PPARGC1B, HDAC4, FAM208B, DOCK8, and SYNE2 reached global significance, and other loci, including MIR146A and OBFC1, reached a suggestive level. Overall, we conclude that most nevus genes affect melanoma risk (KITLG an exception), while many melanoma risk loci do not alter nevus count. For example, variants in TERC and OBFC1 affect both traits, but other telomere length maintenance genes seem to affect melanoma risk only. Our findings implicate multiple pathways in nevogenesis.
Collapse
Affiliation(s)
- David L Duffy
- QIMR Berghofer Medical Research Institute, Brisbane, Australia.
| | - Gu Zhu
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Xin Li
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, 63110, USA
| | - Marianna Sanna
- Department of Twin Research & Genetic Epidemiology, St Thomas Hospital Campus, Kings College, London, UK
| | - Mark M Iles
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Leonie C Jacobs
- Department of Dermatology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - David M Evans
- MRC Integrative Epidemiology Unit, University of Bristol, Bristol, UK
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Seyhan Yazar
- Centre for Ophthalmology and Vision Science, University of Western Australia and the Lions Eye Institute, Perth, Australia
| | | | - Matthew H Law
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Peter Kraft
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Alessia Visconti
- Department of Twin Research & Genetic Epidemiology, St Thomas Hospital Campus, Kings College, London, UK
| | - John C Taylor
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Fan Liu
- Department of Genetic Identification, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | | | - Anjali K Henders
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - Lisa Bowdler
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
| | - Dan Glass
- Department of Twin Research & Genetic Epidemiology, St Thomas Hospital Campus, Kings College, London, UK
| | - M Arfan Ikram
- Department of Epidemiology, Erasmus MC, Rotterdam, Netherlands
| | - André G Uitterlinden
- Department of Epidemiology, Erasmus MC, Rotterdam, Netherlands
- Department of Internal Medicine, Erasmus MC, Rotterdam, Netherlands
| | - Pamela A Madden
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Andrew C Heath
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Elliot C Nelson
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, 63110, USA
| | - Adele C Green
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Molecular Oncology Group, CRUK Manchester Institute, University of Manchester, Manchester, UK
| | - Stephen Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Jennifer H Barrett
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Matthew A Brown
- University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | | | | | - Richard A Sturm
- Dermatology Research Centre, University of Queensland Diamantina Institute, Translational Research Institute, Brisbane, Australia
| | - Alex W Hewitt
- Centre for Ophthalmology and Vision Science, University of Western Australia and the Lions Eye Institute, Perth, Australia
| | - Manfred Kayser
- Department of Genetic Identification, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - David J Hunter
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, 02115, MA, USA
| | - Julia A Newton Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Timothy D Spector
- Department of Twin Research & Genetic Epidemiology, St Thomas Hospital Campus, Kings College, London, UK
| | - Grant W Montgomery
- QIMR Berghofer Medical Research Institute, Brisbane, Australia
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, Australia
| | - David A Mackey
- Centre for Ophthalmology and Vision Science, University of Western Australia and the Lions Eye Institute, Perth, Australia
| | | | - Tamar E Nijsten
- Department of Dermatology, Erasmus MC, University Medical Centre Rotterdam, Rotterdam, The Netherlands
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, UK
| | - Veronique Bataille
- Department of Twin Research & Genetic Epidemiology, St Thomas Hospital Campus, Kings College, London, UK
| | - Mario Falchi
- Department of Twin Research & Genetic Epidemiology, St Thomas Hospital Campus, Kings College, London, UK
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Melvin and Bren Simon Cancer Center, Indiana University, Indianapolis, IN, 63110, USA
| | | |
Collapse
|
25
|
Thomas NE, Edmiston SN, Orlow I, Kanetsky PA, Luo L, Gibbs DC, Parrish EA, Hao H, Busam KJ, Armstrong BK, Kricker A, Cust AE, Anton-Culver H, Gruber SB, Gallagher RP, Zanetti R, Rosso S, Sacchetto L, Dwyer T, Ollila DW, Begg CB, Berwick M, Conway K. Inherited Genetic Variants Associated with Melanoma BRAF/NRAS Subtypes. J Invest Dermatol 2018; 138:2398-2404. [PMID: 29753029 PMCID: PMC6200630 DOI: 10.1016/j.jid.2018.04.025] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 04/08/2018] [Indexed: 10/16/2022]
Abstract
BRAF and NRAS mutations arise early in melanoma development, but their associations with low-penetrance melanoma susceptibility loci remain unknown. In the Genes, Environment and Melanoma Study, 1,223 European-origin participants had their incident invasive primary melanomas screened for BRAF/NRAS mutations and germline DNA genotyped for 47 single-nucleotide polymorphisms identified as low-penetrant melanoma-risk variants. We used multinomial logistic regression to simultaneously examine each single-nucleotide polymorphism's relationship to BRAF V600E, BRAF V600K, BRAF other, and NRAS+ relative to BRAF-/NRAS- melanoma adjusted for study features. IRF4 rs12203592*T was associated with BRAF V600E (odds ratio [OR] = 0.59, 95% confidence interval [CI] = 0.43-0.79) and V600K (OR = 0.65, 95% CI = 0.41-1.03), but not BRAF other or NRAS+ melanoma. A global test of etiologic heterogeneity (Pglobal = 0.001) passed false discovery (Pglobal = 0.0026). PLA2G6 rs132985*T was associated with BRAF V600E (OR = 1.32, 95% CI = 1.05-1.67) and BRAF other (OR = 1.82, 95% CI = 1.11-2.98), but not BRAF V600K or NRAS+ melanoma. The test for etiologic heterogeneity (Pglobal) was 0.005. The IRF4 rs12203592 associations were slightly attenuated after adjustment for melanoma-risk phenotypes. The PLA2G6 rs132985 associations were independent of phenotypes. IRF4 and PLA2G6 inherited genotypes may influence melanoma BRAF/NRAS subtype development.
Collapse
Affiliation(s)
- Nancy E Thomas
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA.
| | - Sharon N Edmiston
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, Moffitt Cancer Center and Research Institute, Tampa, Florida, USA
| | - Li Luo
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - David C Gibbs
- Department of Epidemiology, Emory University, Atlanta, Georgia, USA
| | - Eloise A Parrish
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Honglin Hao
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Klaus J Busam
- Department of Pathology, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Bruce K Armstrong
- School of Public and Global Health, The University of Western Australia, Perth, Australia
| | - Anne Kricker
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Anne E Cust
- Sydney School of Public Health, The University of Sydney, Sydney, Australia; Melanoma Institute Australia, The University of Sydney, North Sydney, Australia
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California, Irvine, California, USA
| | - Stephen B Gruber
- USC Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, USA
| | - Richard P Gallagher
- British Columbia Cancer and Department of Dermatology and Skin Science, University of British Columbia, Vancouver, British Columbia, Canada
| | - Roberto Zanetti
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Stefano Rosso
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy
| | - Lidia Sacchetto
- Piedmont Cancer Registry, Centre for Epidemiology and Prevention in Oncology in Piedmont, Turin, Italy; Politecnico di Torino, Turin, Italy
| | - Terence Dwyer
- George Institute for Global Health, Nuffield Department of Obstetrics and Gynecology, University of Oxford, Oxford, UK
| | - David W Ollila
- Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Surgery, University of North Carolina, Chapel Hill, North Carolina, USA
| | - Colin B Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan-Kettering Cancer Center, New York, USA
| | - Marianne Berwick
- Department of Internal Medicine, University of New Mexico Cancer Center, University of New Mexico, Albuquerque, New Mexico, USA
| | - Kathleen Conway
- Department of Dermatology, University of North Carolina, Chapel Hill, North Carolina, USA; Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, North Carolina, USA; Department of Epidemiology, University of North Carolina, Chapel Hill, North Carolina, USA
| |
Collapse
|
26
|
Zhang T, Choi J, Kovacs MA, Shi J, Xu M, Goldstein AM, Trower AJ, Bishop DT, Iles MM, Duffy DL, MacGregor S, Amundadottir LT, Law MH, Loftus SK, Pavan WJ, Brown KM. Cell-type-specific eQTL of primary melanocytes facilitates identification of melanoma susceptibility genes. Genome Res 2018; 28:1621-1635. [PMID: 30333196 PMCID: PMC6211648 DOI: 10.1101/gr.233304.117] [Citation(s) in RCA: 53] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 09/21/2018] [Indexed: 12/18/2022]
Abstract
Most expression quantitative trait locus (eQTL) studies to date have been performed in heterogeneous tissues as opposed to specific cell types. To better understand the cell-type-specific regulatory landscape of human melanocytes, which give rise to melanoma but account for <5% of typical human skin biopsies, we performed an eQTL analysis in primary melanocyte cultures from 106 newborn males. We identified 597,335 cis-eQTL SNPs prior to linkage disequilibrium (LD) pruning and 4997 eGenes (FDR < 0.05). Melanocyte eQTLs differed considerably from those identified in the 44 GTEx tissue types, including skin. Over a third of melanocyte eGenes, including key genes in melanin synthesis pathways, were unique to melanocytes compared to those of GTEx skin tissues or TCGA melanomas. The melanocyte data set also identified trans-eQTLs, including those connecting a pigmentation-associated functional SNP with four genes, likely through cis-regulation of IRF4 Melanocyte eQTLs are enriched in cis-regulatory signatures found in melanocytes as well as in melanoma-associated variants identified through genome-wide association studies. Melanocyte eQTLs also colocalized with melanoma GWAS variants in five known loci. Finally, a transcriptome-wide association study using melanocyte eQTLs uncovered four novel susceptibility loci, where imputed expression levels of five genes (ZFP90, HEBP1, MSC, CBWD1, and RP11-383H13.1) were associated with melanoma at genome-wide significant P-values. Our data highlight the utility of lineage-specific eQTL resources for annotating GWAS findings, and present a robust database for genomic research of melanoma risk and melanocyte biology.
Collapse
Affiliation(s)
- Tongwu Zhang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jiyeon Choi
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Michael A Kovacs
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Jianxin Shi
- Biostatistics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Mai Xu
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Alisa M Goldstein
- Clinical Genetics Branch, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Adam J Trower
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - Mark M Iles
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, University of Leeds, Leeds, LS9 7TF, United Kingdom
| | - David L Duffy
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Stuart MacGregor
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Laufey T Amundadottir
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Matthew H Law
- Statistical Genetics, QIMR Berghofer Medical Research Institute, Brisbane, Queensland, 4006, Australia
| | - Stacie K Loftus
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20892, USA
| |
Collapse
|
27
|
Tian L, Hu X, He Y, Wu Z, Li D, Zhang H. Construction of lncRNA-miRNA-mRNA networks reveals functional lncRNAs in abdominal aortic aneurysm. Exp Ther Med 2018; 16:3978-3986. [PMID: 30344676 PMCID: PMC6176170 DOI: 10.3892/etm.2018.6690] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2017] [Accepted: 03/01/2018] [Indexed: 01/12/2023] Open
Abstract
Abdominal aortic aneurysm (AAA) is one of the most significant causes of morbidity and mortality in populations aged >65 years worldwide. However, the underlying mechanisms of AAA based on the competitive endogenous RNA (ceRNA) hypothesis have remained elusive. In the present study, differently expressed long non-coding RNA (lncRNA)-microRNA (miRNA)-mRNA networks in AAA were constructed by analyzing public datasets, including GSE7084, GSE24194 from rats and that of a previous study. A total of 1,219 mRNAs, 2,093 lncRNAs and 57 miRNAs were identified to differently express in AAA. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes analyses were performed to explore the potential roles of differently expressed lncRNAs based on their regulating mRNAs. Based on the ceRNA hypothesis, lncRNA-miRNA-mRNA networks in AAA were, for the first time, constructed at a system-wide level. The present study identified 5 upregulated lncRNAs [nuclear paraspeckle assembly transcript 1, cyclin-dependent kinase inhibitor 2B antisense RNA 1, small Cajal body-specific RNA 10, AC005224.4 and SUMO1/sentrin/SMT3-specific peptidase 3-eukaryotic translation initiation factor 4A1] and the downregulated zinc ribbon domain containing 1 antisense RNA 1 as key lncRNAs in ceRNA networks. To the best of our knowledge, the present study was the first to screen ceRNA networks in AAA. In addition, key lncRNA-mRNA-biological processes analysis indicated that these key lncRNAs were involved in regulating signal transduction, protein amino acid phosphorylation, immune response, transcription, development and cell differentiation. The present study provides novel clues to explore the molecular mechanisms of AAA progression in terms of lncRNA implication.
Collapse
Affiliation(s)
- Lu Tian
- Department of Vascular Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Xiaofeng Hu
- Department of Cardiology, Zhejiang Hospital, Hangzhou, Zhejiang 310013, P.R. China
| | - Yangyan He
- Department of Vascular Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Ziheng Wu
- Department of Vascular Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Donglin Li
- Department of Vascular Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| | - Hongkun Zhang
- Department of Vascular Surgery, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
28
|
Stucci LS, D'Oronzo S, Tucci M, Macerollo A, Ribero S, Spagnolo F, Marra E, Picasso V, Orgiano L, Marconcini R, De Rosa F, Di Guardo L, Galli G, Gandini S, Palmirotta R, Palmieri G, Queirolo P, Silvestris F. Vitamin D in melanoma: Controversies and potential role in combination with immune check-point inhibitors. Cancer Treat Rev 2018; 69:21-28. [PMID: 29864718 DOI: 10.1016/j.ctrv.2018.05.016] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 05/29/2018] [Accepted: 05/30/2018] [Indexed: 12/17/2022]
Abstract
The role of vitamin D in melanoma is still controversial. Although several Authors described a correlation between vitamin D deficiency and poor survival in metastatic melanoma patients, clinical trials exploring the effects of vitamin D supplementation in this clinical setting were mostly inconclusive. However, recent evidence suggests that vitamin D exerts both anti-proliferative effects on tumor cells and immune-modulating activities, that have been widely explored in auto-immune disorders. On the one hand, vitamin D has been shown to inhibit T-helper17 lymphocytes, notoriously involved in the pathogenesis of immune-related adverse events (iAEs) which complicate immune-checkpoint inhibitor (ICI) treatment. On the other hand, vitamin D up-regulates PDL-1 expression on both epithelial and immune cells, suggesting a synergic effect in combination with ICIs, for which further investigation is needed.
Collapse
Affiliation(s)
- Luigia Stefania Stucci
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Italy
| | - Stella D'Oronzo
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Italy.
| | - Marco Tucci
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Italy
| | - Antonella Macerollo
- Sobell Department of Motor Neuroscience and Movement Disorders, UCL Institute of Neurology, London WC1N 3BG, United Kingdom
| | - Simone Ribero
- Department of Medical Sciences Section of Dermatology, University of Turin, Italy
| | - Francesco Spagnolo
- Department of Medical Oncology , Ospedale Policlinico San Martino, Genova, Italy
| | - Elena Marra
- Department of Medical Sciences Section of Dermatology, University of Turin, Italy
| | - Virginia Picasso
- Department of Medical Oncology , Ospedale Policlinico San Martino, Genova, Italy
| | - Laura Orgiano
- Department of Medical Oncology, University of Cagliari, Cagliari, Italy
| | - Riccardo Marconcini
- Department of Oncology, Azienda Ospedaliero-Universitaria Pisana and University of Pisa, Istituto Toscano Tumori, Santa Chiara Hospital, Pisa, Italy
| | - Francesco De Rosa
- Immunotherapy-Cell Therapy and Biobank Unit of Biostatistics and Clinical Trials, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori (IRST) IRCCS, Meldola, Italy
| | - Lorenza Di Guardo
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Giulia Galli
- Department of Medical Oncology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
| | - Sara Gandini
- Division of Epidemiology and Biostatistics, European Institute of Oncology, Milan, Italy
| | - Raffaele Palmirotta
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Italy
| | | | - Paola Queirolo
- Department of Medical Oncology , Ospedale Policlinico San Martino, Genova, Italy
| | - Francesco Silvestris
- Medical Oncology Unit, Department of Biomedical Sciences and Human Oncology, University of Bari 'Aldo Moro', Italy
| |
Collapse
|
29
|
Bourneuf E, Estellé J, Blin A, Créchet F, Schneider MDP, Gilbert H, Brossard M, Vaysse A, Lathrop M, Vincent-Naulleau S, Demenais F. New susceptibility loci for cutaneous melanoma risk and progression revealed using a porcine model. Oncotarget 2018; 9:27682-27697. [PMID: 29963229 PMCID: PMC6021234 DOI: 10.18632/oncotarget.25455] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 05/05/2018] [Indexed: 01/08/2023] Open
Abstract
Despite major advances, it is estimated that a large part of melanoma predisposing genes remains to be discovered. Animal models of spontaneous diseases are valuable tools and experimental crosses can be used to identify and fine-map new susceptibility loci associated with melanoma. We performed a Genome-Wide Association Study (GWAS) of melanoma occurrence and progression (clinical ulceration and presence of metastasis) in a porcine model of spontaneous melanoma, the MeLiM pig. Five loci on chromosomes 2, 5, 7, 8 and 16 showed genome-wide significant associations (p < 5 × 10–6) with either one of these phenotypes. Suggestive associations (p < 5 × 10–5) were also found at 16 additional loci. Moreover, comparison of the porcine results to those reported by human melanoma GWAS indicated shared association signals notably at CDKAL1 and TERT loci but also nearby CCND1, FTO, PLA2G6 and TMEM38B-RAD23B loci. Extensive search of the literature revealed a potential key role of genes at the identified porcine loci in tumor invasion (DST, PLEKHA5, CBY1, LIMK2 and ETV5) and immune response modulation (ETV5, HERC3 and DICER1) of the progression phenotypes. These biological processes are consistent with the clinico-pathological features of MeLiM tumors and can open new routes for future melanoma research in humans.
Collapse
Affiliation(s)
- Emmanuelle Bourneuf
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Jordi Estellé
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Amandine Blin
- GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Outils et Méthodes de la Systématique Intégrative, OMSI-UMS 2700, CNRS MNHN, Muséum National d'Histoire Naturelle, Paris, France
| | - Françoise Créchet
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Maria Del Pilar Schneider
- GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France.,Present address: Ipsen Innovation, Les Ulis, France
| | - Hélène Gilbert
- GenPhyse, INRA, Université de Toulouse, INPT, ENVT, Castanet Tolosan, France
| | - Myriam Brossard
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Amaury Vaysse
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| | - Mark Lathrop
- McGill University and Genome Québec Innovation Centre, Montréal, Québec, Canada
| | - Silvia Vincent-Naulleau
- CEA, DRF/iRCM/SREIT/LREG, Jouy-en-Josas, France.,GABI, INRA, AgroParisTech, Université Paris-Saclay, Jouy-en-Josas, France
| | - Florence Demenais
- INSERM, UMR-946, Genetic Variation and Human Diseases Unit, Paris, France.,Institut Universitaire d'Hématologie, Université Paris Diderot, Sorbonne Paris Cité, Paris, France
| |
Collapse
|
30
|
Snetselaar R, van Oosterhout MFM, Grutters JC, van Moorsel CHM. Telomerase Reverse Transcriptase Polymorphism rs2736100: A Balancing Act between Cancer and Non-Cancer Disease, a Meta-Analysis. Front Med (Lausanne) 2018. [PMID: 29536006 PMCID: PMC5835035 DOI: 10.3389/fmed.2018.00041] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The enzyme telomerase reverse transcriptase (TERT) is essential for telomere maintenance. In replicating cells, maintenance of telomere length is important for the preservation of vital genetic information and prevention of genomic instability. A common genetic variant in TERT, rs2736100 C/A, is associated with both telomere length and multiple diseases. Carriage of the C allele is associated with longer telomere length, while carriage of the A allele is associated with shorter telomere length. Furthermore, some diseases have a positive association with the C and some with the A allele. In this study, meta-analyses were performed for two groups of diseases, cancerous diseases, e.g., lung cancer and non-cancerous diseases, e.g., pulmonary fibrosis, using data from genome-wide association studies and case-control studies. In the meta-analysis it was found that cancer positively associated with the C allele (pooled OR 1.16 [95% CI 1.09–1.23]) and non-cancerous diseases negatively associated with the C allele (pooled OR 0.81 [95% CI 0.65–0.99]). This observation illustrates that the ambiguous role of telomere maintenance in disease hinges, at least in part, on a single locus in telomerase genes. The dual role of this single nucleotide polymorphism also emphasizes that therapeutic agents aimed at influencing telomere maintenance should be used with caution.
Collapse
Affiliation(s)
- Reinier Snetselaar
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, Netherlands
| | - Matthijs F M van Oosterhout
- Interstitial Lung Diseases Center of Excellence, Department of Pathology, St Antonius Hospital, Nieuwegein, Netherlands
| | - Jan C Grutters
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, Netherlands.,Division of Heart and Lung, University Medical Center Utrecht, Utrecht, Netherlands
| | - Coline H M van Moorsel
- Interstitial Lung Diseases Center of Excellence, Department of Pulmonology, St Antonius Hospital, Nieuwegein, Netherlands.,Division of Heart and Lung, University Medical Center Utrecht, Utrecht, Netherlands
| |
Collapse
|
31
|
Benfodda M, Gazal S, Descamps V, Basset-Seguin N, Deschamps L, Thomas L, Lebbe C, Saiag P, Zanetti R, Sacchetto L, Chiorino G, Scatolini M, Grandchamp B, Bensussan A, Soufir N. Truncating mutations of TP53AIP1 gene predispose to cutaneous melanoma. Genes Chromosomes Cancer 2018; 57:294-303. [PMID: 29359367 DOI: 10.1002/gcc.22528] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2017] [Revised: 01/15/2018] [Accepted: 01/18/2018] [Indexed: 01/08/2023] Open
Abstract
Genetic predisposition to cutaneous malignant melanoma (CMM) involves highly penetrant predisposing genes and low and intermediate penetrant predisposing alleles. However, the missing heritability in (CMM) is still high. For such and in order to identify new genetic factors for CMM, we conducted an exome sequencing study in high-risk CMM patients. Two rounds of exome sequencing were successively performed in 33 and 27 high-risk patients. We focused on genes carrying rare nonsense, frameshift, and splice variants (allelic frequency <1%) that were present in both series of exomes. An extension study was then conducted in a large cohort (1 079 CMM patients and 1 230 Caucasian ethnically matched healthy controls), and the inactivating variants frequency was compared between groups using two-sided Fisher exact test. Two TP53AIP1 truncating mutations were identified in four patients: a frameshift c.63_64insG, p.Q22Afs*81 in two patients from the same family and in the proband of a second family; and a nonsense mutation c.95 C > A, p.Ser32Stop in a patient with multiple CMMs. In all patients, TP53AIP1 truncating variants were strongly associated with CMM risk (two-sided Fisher exact test = 0.004, OR = 3.3[1.3-8.5]). Additionally, we showed that TP53AIP1 mRNA was strongly down-regulated throughout different phases of melanoma progression. TP53AIP1 gene is a TP53 target which plays a key role by inducting apoptosis in response to UV-induced DNA damage. Constitutional mutations of TP53AIP1 had previously been involved in susceptibility to prostate cancer. Our results show that constitutional truncating TP53AIP1 mutations predispose to CMM in the French population. Replication studies in other populations should be performed.
Collapse
Affiliation(s)
- Meriem Benfodda
- INSERM U976, Centre de Recherche sur la Peau, Hôpital Saint Louis, 75010, Paris, France.,Département de Génétique, Hôpital Bichat Claude Bernard, APHP, 75018, Paris, France
| | - Steven Gazal
- Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France.,UMR S 738, Faculté de Médecine Xavier Bichat, 75018, Paris, France
| | - Vincent Descamps
- INSERM U976, Centre de Recherche sur la Peau, Hôpital Saint Louis, 75010, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France.,Département de Dermatologie, Hôpital Bichat Claude Bernard, APHP, 75018, Paris, France
| | - Nicole Basset-Seguin
- INSERM U976, Centre de Recherche sur la Peau, Hôpital Saint Louis, 75010, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France.,Département de Dermatologie, Hôpital Saint Louis, APHP, 75010, Paris, France
| | - Lydia Deschamps
- Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France.,Département d'Anatomie Pathologique, Hôpital Bichat Claude Bernard, APHP, 75018, Paris, France
| | - Luc Thomas
- Département de Dermatologie, Hôpital de l'Hôtel-Dieu, 69002, Lyon, France
| | - Celeste Lebbe
- INSERM U976, Centre de Recherche sur la Peau, Hôpital Saint Louis, 75010, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France.,Département de Dermatologie, Hôpital Saint Louis, APHP, 75010, Paris, France
| | - Philippe Saiag
- Département de Dermatologie, Hôpital Ambroise Paré, APHP, 92100, Boulogne Billancourt, France
| | - Roberto Zanetti
- Centre for Cancer Prevention, Piedmont Cancer Registry-CPO, Torino, Italy
| | - Lidia Sacchetto
- Centre for Cancer Prevention, Piedmont Cancer Registry-CPO, Torino, Italy.,Politecnico di Torino, Torino, Italy.,Università degli Studi di Torino, Torino, Italy.,Department of Mathematical Sciences, Politecnico di Torino, Torino, Italy
| | - Giovanna Chiorino
- Centre for Cancer Prevention, Piedmont Cancer Registry-CPO, Torino, Italy
| | - Maria Scatolini
- Laboratory of Molecular Oncology, Fondazione Edo ed Elvo Tempia, Biella, Italy
| | - Bernard Grandchamp
- Département de Génétique, Hôpital Bichat Claude Bernard, APHP, 75018, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France
| | - Armand Bensussan
- INSERM U976, Centre de Recherche sur la Peau, Hôpital Saint Louis, 75010, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France
| | - Nadem Soufir
- INSERM U976, Centre de Recherche sur la Peau, Hôpital Saint Louis, 75010, Paris, France.,Département de Génétique, Hôpital Bichat Claude Bernard, APHP, 75018, Paris, France.,Université Paris Diderot, Sorbonne Paris Cité, 75005, Paris, France
| |
Collapse
|
32
|
Bose A, Petsko GA, Eliezer D. Parkinson's Disease and Melanoma: Co-Occurrence and Mechanisms. JOURNAL OF PARKINSON'S DISEASE 2018; 8:385-398. [PMID: 29991141 PMCID: PMC6130416 DOI: 10.3233/jpd-171263] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 04/20/2018] [Indexed: 12/31/2022]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder that is characterized by loss of dopaminergic neurons in the substantia nigra pars compacta, depletion of dopamine in the striatum and the presence of Lewy bodies. Cancer is uncontrolled growth of cells in the body and migration of these cells from their site of origin to other parts of the body. PD and cancer are two opposite diseases, one arising from cell proliferation and the other from cell degeneration. This fundamental difference is consistent with inverse comorbidity between most cancers and neurodegenerative diseases. However, a positive association of PD and melanoma has been reported which has recently become of significant interest. A link between PD and cancer has been supported by many epidemiological studies, most of which show that PD patients have a lower risk of developing most cancers than the general population. However, the mechanisms underlying this epidemiological observation are not known. In this review we focus on epidemiological studies correlating PD and melanoma and the possible mechanisms underlying the co-occurrence of the two diseases. We explore possible explanations for the important observations that more PD patients develop melanoma that would otherwise be expected and vice-versa.
Collapse
Affiliation(s)
- Anindita Bose
- Helen and Robert Appel Alzheimer’s disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Gregory A. Petsko
- Helen and Robert Appel Alzheimer’s disease Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - David Eliezer
- Department of Biochemistry, Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| |
Collapse
|
33
|
Goldstein AM, Xiao Y, Sampson J, Zhu B, Rotunno M, Bennett H, Wen Y, Jones K, Vogt A, Burdette L, Luo W, Zhu B, Yeager M, Hicks B, Han J, De Vivo I, Koutros S, Andreotti G, Beane-Freeman L, Purdue M, Freedman ND, Chanock SJ, Tucker MA, Yang XR. Rare germline variants in known melanoma susceptibility genes in familial melanoma. Hum Mol Genet 2017; 26:4886-4895. [PMID: 29036293 PMCID: PMC5886297 DOI: 10.1093/hmg/ddx368] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2017] [Revised: 09/11/2017] [Accepted: 09/19/2017] [Indexed: 12/20/2022] Open
Abstract
Known high-risk cutaneous malignant melanoma (CMM) genes account for melanoma risk in <40% of melanoma-prone families, suggesting the existence of additional high-risk genes or perhaps a polygenic mechanism involving multiple genetic modifiers. The goal of this study was to systematically characterize rare germline variants in 42 established melanoma genes among 144 CMM patients in 76 American CMM families without known mutations using data from whole-exome sequencing. We identified 68 rare (<0.1% in public and in-house control datasets) nonsynonymous variants in 25 genes. We technically validated all loss-of-function, inframe insertion/deletion, and missense variants predicted as deleterious, and followed them up in 1, 559 population-based CMM cases and 1, 633 controls. Several of these variants showed disease co-segregation within families. Of particular interest, a stopgain variant in TYR was present in five of six CMM cases/obligate gene carriers in one family and a single population-based CMM case. A start gain variant in the 5'UTR region of PLA2G6 and a missense variant in ATM were each seen in all three affected people in a single family, respectively. Results from rare variant burden tests showed that familial and population-based CMM patients tended to have higher frequencies of rare germline variants in albinism genes such as TYR, TYRP1, and OCA2 (P < 0.05). Our results suggest that rare nonsynonymous variants in low- or intermediate-risk CMM genes may influence familial CMM predisposition, warranting further investigation of both common and rare variants in genes affecting functionally important pathways (such as melanogenesis) in melanoma risk assessment.
Collapse
Affiliation(s)
- Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Yanzi Xiao
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Joshua Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Melissa Rotunno
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Hunter Bennett
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Yixuan Wen
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Kristine Jones
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Aurelie Vogt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Laurie Burdette
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Wen Luo
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Bin Zhu
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Meredith Yeager
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Belynda Hicks
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
- Cancer Genomics Research Laboratory, Leidos Biomedical Research, Frederick National Laboratory for Cancer Research, Frederick, MD, USA
| | - Jiali Han
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Melvin & Bren Simon Cancer Center, Indiana University, Indianapolis, IN, USA
| | - Immaculata De Vivo
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Stella Koutros
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Gabriella Andreotti
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Laura Beane-Freeman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Mark Purdue
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Neal D Freedman
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| | - Xiaohong R Yang
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, NIH, DHHS, Bethesda, MD, USA
| |
Collapse
|
34
|
Taylor NJ, Mitra N, Goldstein AM, Tucker MA, Avril MF, Azizi E, Bergman W, Bishop DT, Bressac-de Paillerets B, Bruno W, Calista D, Cannon-Albright LA, Cuellar F, Cust AE, Demenais F, Elder DE, Gerdes AM, Ghiorzo P, Grazziotin TC, Hansson J, Harland M, Hayward NK, Hocevar M, Höiom V, Ingvar C, Landi MT, Landman G, Larre-Borges A, Leachman SA, Mann GJ, Nagore E, Olsson H, Palmer JM, Perić B, Pjanova D, Pritchard A, Puig S, van der Stoep N, Wadt KAW, Whitaker L, Yang XR, Newton Bishop JA, Gruis NA, Kanetsky PA. Germline Variation at CDKN2A and Associations with Nevus Phenotypes among Members of Melanoma Families. J Invest Dermatol 2017; 137:2606-2612. [PMID: 28830827 PMCID: PMC5701856 DOI: 10.1016/j.jid.2017.07.829] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2017] [Revised: 07/21/2017] [Accepted: 07/30/2017] [Indexed: 11/17/2022]
Abstract
Germline mutations in CDKN2A are frequently identified among melanoma kindreds and are associated with increased atypical nevus counts. However, a clear relationship between pathogenic CDKN2A mutation carriage and other nevus phenotypes including counts of common acquired nevi has not yet been established. Using data from GenoMEL, we investigated the relationships between CDKN2A mutation carriage and 2-mm, 5-mm, and atypical nevus counts among blood-related members of melanoma families. Compared with individuals without a pathogenic mutation, those who carried one had an overall higher prevalence of atypical (odds ratio = 1.64; 95% confidence interval = 1.18-2.28) nevi but not 2-mm nevi (odds ratio = 1.06; 95% confidence interval = 0.92-1.21) or 5-mm nevi (odds ratio = 1.26; 95% confidence interval = 0.94-1.70). Stratification by case status showed more pronounced positive associations among non-case family members, who were nearly three times (odds ratio = 2.91; 95% confidence interval = 1.75-4.82) as likely to exhibit nevus counts at or above the median in all three nevus categories simultaneously when harboring a pathogenic mutation (vs. not harboring one). Our results support the hypothesis that unidentified nevogenic genes are co-inherited with CDKN2A and may influence carcinogenesis.
Collapse
Affiliation(s)
- Nicholas J Taylor
- Department of Epidemiology and Biostatistics, Texas A&M Health Science Center, College Station, Texas, USA
| | - Nandita Mitra
- Department of Biostatistics and Epidemiology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Alisa M Goldstein
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Margaret A Tucker
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Marie-Françoise Avril
- Assistance Publique-Hôpitaux de Paris, Hôpital Cochin et Université Paris Descartes, Paris, France
| | - Esther Azizi
- Department of Dermatology, Sheba Medical Center, Tel Hashomer, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Wilma Bergman
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - D Timothy Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Cancer Research UK Clinical Centre at Leeds, St James's University Hospital, Leeds, UK
| | - Brigitte Bressac-de Paillerets
- Gustave Roussy, Université Paris-Saclay, Département de Biologie et Pathologie Médicales, INSERM, U1186, Villejuif, France
| | - William Bruno
- Department of Internal Medicine and Medical Specialties, University of Genoa and IRCCS AOU San Martino-IST Genoa, Italy
| | - Donato Calista
- Dermatology Unit, Maurizio Bufalini Hospital, Cesena, Italy
| | - Lisa A Cannon-Albright
- Departments of Genetic Epidemiology and Biomedical Informatics, University of Utah, Salt Lake City, Utah, USA
| | - Francisco Cuellar
- Melanoma Unit, Dermatology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain; CIBER de Enfermedades Raras, Barcelona, Spain
| | - Anne E Cust
- Sydney School of Public Health, University of Sydney, Sydney, New South Wales, Australia; Melanoma Institute Australia, Westmead, New South Wales, Australia
| | - Florence Demenais
- Genetic Variation and Human Diseases Unit, UMR-946, INSERM, Université Paris Diderot, Université Sorbonne Paris Cité, Paris, France
| | - David E Elder
- Departments of Pathology and Laboratory Medicine, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Anne-Marie Gerdes
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Paola Ghiorzo
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Cancer Research UK Clinical Centre at Leeds, St James's University Hospital, Leeds, UK
| | - Thais C Grazziotin
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Rio Grande do Sul, Brazil
| | - Johan Hansson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Mark Harland
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Cancer Research UK Clinical Centre at Leeds, St James's University Hospital, Leeds, UK
| | - Nicholas K Hayward
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Marko Hocevar
- Institute of Oncology Ljubljana, Zaloska, Ljubljana, Slovenia
| | - Veronica Höiom
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Christian Ingvar
- Departments of Clinical Sciences and Surgery, Lund University, Lund, Sweden
| | - Maria Teresa Landi
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Gilles Landman
- Department of Pathology, Escola Paulista de Medicina, UNIFESP, São Paulo, Brazil
| | - Alejandra Larre-Borges
- Unidad de Lesiones Pigmentadas, Cátedra de Dermatología, Hospital de Clínicas, Universidad de la República, Montevideo, Uruguay
| | - Sancy A Leachman
- Department of Dermatology, Oregon Health & Science University, Portland, Oregon, USA
| | - Graham J Mann
- Melanoma Institute Australia, Westmead, New South Wales, Australia; Westmead Institute for Cancer Research, University of Sydney at Westmead Millennium Institute, New South Wales, Australia
| | - Eduardo Nagore
- Department of Dermatology, Instituto Valenciano de Oncologia, Valencia, Spain
| | - Håkan Olsson
- Departments of Clinical Sciences and Surgery, Lund University, Lund, Sweden
| | - Jane M Palmer
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Barbara Perić
- Institute of Oncology Ljubljana, Zaloska, Ljubljana, Slovenia
| | - Dace Pjanova
- Latvian Biomedical Research and Study Centre, Riga, Latvia
| | - Antonia Pritchard
- QIMR Berghofer Medical Research Institute, Herston, Queensland, Australia
| | - Susana Puig
- Melanoma Unit, Dermatology Department, Hospital Clinic, IDIBAPS, Barcelona, Spain; CIBER de Enfermedades Raras, Barcelona, Spain
| | - Nienke van der Stoep
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Karin A W Wadt
- Department of Clinical Genetics, University Hospital of Copenhagen, Copenhagen, Denmark
| | - Linda Whitaker
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Cancer Research UK Clinical Centre at Leeds, St James's University Hospital, Leeds, UK
| | - Xiaohong R Yang
- Human Genetics Program, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, Maryland, USA
| | - Julia A Newton Bishop
- Section of Epidemiology and Biostatistics, Leeds Institute of Cancer and Pathology, Cancer Research UK Clinical Centre at Leeds, St James's University Hospital, Leeds, UK
| | - Nelleke A Gruis
- Department of Dermatology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Peter A Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida, USA.
| |
Collapse
|
35
|
|
36
|
Melanocytic nevi and melanoma: unraveling a complex relationship. Oncogene 2017; 36:5771-5792. [PMID: 28604751 DOI: 10.1038/onc.2017.189] [Citation(s) in RCA: 129] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2017] [Revised: 05/09/2017] [Accepted: 05/10/2017] [Indexed: 12/11/2022]
Abstract
Approximately 33% of melanomas are derived directly from benign, melanocytic nevi. Despite this, the vast majority of melanocytic nevi, which typically form as a result of BRAFV600E-activating mutations, will never progress to melanoma. Herein, we synthesize basic scientific insights and data from mouse models with common observations from clinical practice to comprehensively review melanocytic nevus biology. In particular, we focus on the mechanisms by which growth arrest is established after BRAFV600E mutation. Means by which growth arrest can be overcome and how melanocytic nevi relate to melanoma are also considered. Finally, we present a new conceptual paradigm for understanding the growth arrest of melanocytic nevi in vivo termed stable clonal expansion. This review builds upon the canonical hypothesis of oncogene-induced senescence in growth arrest and tumor suppression in melanocytic nevi and melanoma.
Collapse
|
37
|
Armstrong BK, Cust AE. Sun exposure and skin cancer, and the puzzle of cutaneous melanoma: A perspective on Fears et al. Mathematical models of age and ultraviolet effects on the incidence of skin cancer among whites in the United States. American Journal of Epidemiology 1977; 105: 420-427. Cancer Epidemiol 2017; 48:147-156. [PMID: 28478931 DOI: 10.1016/j.canep.2017.04.004] [Citation(s) in RCA: 79] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2017] [Revised: 04/06/2017] [Accepted: 04/08/2017] [Indexed: 11/19/2022]
Abstract
Sunlight has been known as an important cause of skin cancer since around the turn of the 20th Century. A 1977 landmark paper of US scientists Fears, Scotto, and Schneiderman advanced a novel hypothesis whereby cutaneous melanoma was primarily caused by intermittent sun exposure (i.e. periodic, brief episodes of exposure to high-intensity ultraviolet radiation) while the keratinocyte cancers, squamous cell carcinoma and basal cell carcinoma, were primarily caused by progressive accumulation of sun exposure. With respect to cutaneous melanoma, this became known as the intermittent exposure hypothesis. The hypothesis stemmed from analysis of measured ambient ultraviolet radiation and age-specific incidence rates of melanoma and keratinocyte cancers collected as an extension to the US Third National Cancer Survey in several US States. In this perspective paper, we put this novel hypothesis into the context of knowledge at the time, and describe subsequent epidemiological and molecular research into melanoma that elaborated the intermittent exposure hypothesis and ultimately replaced it with a dual pathway hypothesis. Our present understanding is of two distinct biological pathways by which cutaneous melanoma might develop; a nevus prone pathway initiated by early sun exposure and promoted by intermittent sun exposure or possibly host factors; and a chronic sun exposure pathway in sun sensitive people who progressively accumulate sun exposure to the sites of future melanomas.
Collapse
Affiliation(s)
- Bruce K Armstrong
- Cancer Epidemiology and Prevention Research Group, School of Public Health, The University of Sydney, NSW, Australia; School of Global and Population Health, The University of Western Australia, Perth, WA, Australia.
| | - Anne E Cust
- Cancer Epidemiology and Prevention Research Group, School of Public Health, The University of Sydney, NSW, Australia; The Melanoma Institute Australia, North Sydney, NSW, Australia
| |
Collapse
|
38
|
Anbunathan H, Bowcock AM. The Molecular Revolution in Cutaneous Biology: The Era of Genome-Wide Association Studies and Statistical, Big Data, and Computational Topics. J Invest Dermatol 2017; 137:e113-e118. [PMID: 28411841 DOI: 10.1016/j.jid.2016.03.047] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2015] [Revised: 02/10/2016] [Accepted: 03/02/2016] [Indexed: 01/04/2023]
Abstract
The investigation of biological systems involving all organs of the body including the skin is in era of big data. This requires heavy-duty computational tools, and novel statistical methods. Microarrays have allowed the interrogation of thousands of common genetic markers in thousands of individuals from the same population (termed genome wide association studies or GWAS) to reveal common variation associated with disease or phenotype. These markers are usually single nucleotide polymorphisms (SNPs) that are relatively common in the population. In the case of dermatological diseases such as alopecia areata, vitiligo, psoriasis and atopic dermatitis, common variants have been identified that are associated with disease, and these provide insights into biological pathways and reveal possible novel drug targets. Other skin phenotypes such as acne, color and skin cancers are also being investigated with GWAS. Analyses of such large GWAS datasets require a consideration of a number of statistical issues including the testing of multiple markers, population substructure, and ultimately a requirement for replication. There are also issues regarding the missing heritability of disease that cannot be entirely explained with current GWAS approaches. Next generation sequencing technologies such as exome and genome sequencing of similar patient cohorts will reveal additional variants contributing to disease susceptibility. However, the data generated with these approaches will be orders of magnitude greater than that those generated with arrays, with concomitant challenges in the identification of disease causing variants.
Collapse
Affiliation(s)
- Hima Anbunathan
- National Heart and Lung Institute, Imperial College, London, UK
| | - Anne M Bowcock
- National Heart and Lung Institute, Imperial College, London, UK.
| |
Collapse
|
39
|
Cai M, Dai S, Chen W, Xia C, Lu L, Dai S, Qi J, Wang M, Wang M, Zhou L, Lei F, Zuo T, Zeng H, Zhao X. Environmental factors, seven GWAS-identified susceptibility loci, and risk of gastric cancer and its precursors in a Chinese population. Cancer Med 2017; 6:708-720. [PMID: 28220687 PMCID: PMC5345626 DOI: 10.1002/cam4.1038] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 01/16/2017] [Accepted: 01/18/2017] [Indexed: 12/15/2022] Open
Abstract
Gene–environment interactions may increase gastric cancer (GC) risk. Seven susceptibility loci identified by genome‐wide association studies (GWASs) suggest that genetic factors play a role in gastric carcinogenesis. Meanwhile, Helicobacter pylori (H. pylori) infection, smoking, and alcohol drinking are also important environmental factors for gastric cancer. However, studies to explore the role of gene–environment interactions in gastric carcinogenesis, and particularly the relationship between the seven susceptibility loci and their potential interactions with H. pylori infection, smoking, and alcohol drinking in risk of GC, and severe intestinal metaplasia (IM)/dysplasia, have been inconclusive. A total of 1273 subjects in a Chinese population were recruited, and genotyping was carried out using the competitive allele‐specific PCR (KASP) method. Unconditional logistic regression was applied to model the associations between genetic polymorphisms and the disease risk. Effect modifications by H. pylori infection, smoking and alcohol drinking were evaluated. PSCA rs2294008/rs2976392 showed a significant, multiplicative interaction with H. pylori infection in risk of GC. Meanwhile, PRKAA1 rs13361707 had an additive interaction with H. pylori infection. SLC52A3 rs13042395 showed an interaction with alcohol drinking in risk of GC. Moreover, three SNPs, MUC1 rs4072037, ZBTB20 rs9841504 and PRKAA1 rs13361707, were associated with precancerous gastric lesions (severe IM/dysplasia). Our data suggest that genetic predisposition factors identified by GWAS may interact with environmental risk factors, Particularly for H. pylori infection and alcohol consumption, to increase the risk of GC.
Collapse
Affiliation(s)
- Meng Cai
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuyang Dai
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wanqing Chen
- National Office for Cancer Prevention and Control, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Changfa Xia
- National Office for Cancer Prevention and Control, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lingeng Lu
- Department of Chronic Disease Epidemiology, Yale School of Public Health, School of Medicine, Yale Cancer Center, Yale University, New Haven, Connecticut, USA
| | - Shuguang Dai
- Center for Disease Control and Prevention of Sheyang County, Sheyang, Jiangsu, China
| | - Jun Qi
- Department of Clinical Laboratory, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Minjie Wang
- Department of Clinical Laboratory, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Meilin Wang
- Department of Environmental Genomics, Jiangsu Key Laboratory of Cancer Biomarkers, Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing Medical University, Nanjing, Jiangsu, China.,Department of Genetic Toxicology, the Key Laboratory of Modern Toxicology of Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing, Jiangsu, China
| | - Lanping Zhou
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fuhua Lei
- Department of Pathology, Feicheng People Hospital, Feicheng, Shandong, China
| | - Tingting Zuo
- National Office for Cancer Prevention and Control, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongmei Zeng
- National Office for Cancer Prevention and Control, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaohang Zhao
- State Key Laboratory of Molecular Oncology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
40
|
Sangalli A, Malerba G, Tessari G, Rodolfo M, Gomez-Lira M. Melanoma risk alleles are associated with downregulation of the MTAP gene and hypermethylation of a CpG island upstream of the gene in dermal fibroblasts. Exp Dermatol 2017; 26:733-736. [DOI: 10.1111/exd.13247] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2016] [Indexed: 01/23/2023]
Affiliation(s)
- Antonella Sangalli
- Department of Neurosciences, Biomedicine and Movement Sciences; Section of Biology and Genetics; University of Verona; Verona Italy
| | - Giovanni Malerba
- Department of Neurosciences, Biomedicine and Movement Sciences; Section of Biology and Genetics; University of Verona; Verona Italy
| | - Gianpaolo Tessari
- Section of Dermatology and Venereal Diseases; Department of Medicine; University of Verona; Verona Italy
| | - Monica Rodolfo
- Unit of Immunotherapy; Fondazione IRCCS Istituto Nazionale dei Tumori; Milan Italy
| | - Macarena Gomez-Lira
- Department of Neurosciences, Biomedicine and Movement Sciences; Section of Biology and Genetics; University of Verona; Verona Italy
| |
Collapse
|
41
|
MURAKAMI M. Lipoquality control by phospholipase A 2 enzymes. PROCEEDINGS OF THE JAPAN ACADEMY. SERIES B, PHYSICAL AND BIOLOGICAL SCIENCES 2017; 93:677-702. [PMID: 29129849 PMCID: PMC5743847 DOI: 10.2183/pjab.93.043] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2023]
Abstract
The phospholipase A2 (PLA2) family comprises a group of lipolytic enzymes that typically hydrolyze the sn-2 position of glycerophospholipids to give rise to fatty acids and lysophospholipids. The mammalian genome encodes more than 50 PLA2s or related enzymes, which are classified into several subfamilies on the basis of their structures and functions. From a general viewpoint, the PLA2 family has mainly been implicated in signal transduction, producing bioactive lipid mediators derived from fatty acids and lysophospholipids. Recent evidence indicates that PLA2s also contribute to phospholipid remodeling for membrane homeostasis or energy production for fatty acid β-oxidation. Accordingly, PLA2 enzymes can be regarded as one of the key regulators of the quality of lipids, which I herein refer to as lipoquality. Disturbance of PLA2-regulated lipoquality hampers tissue and cellular homeostasis and can be linked to various diseases. Here I overview the current state of understanding of the classification, enzymatic properties, and physiological functions of the PLA2 family.
Collapse
Affiliation(s)
- Makoto MURAKAMI
- Laboratory of Environmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, the University of Tokyo, Tokyo, Japan
- Lipid Metabolism Project, Tokyo Metropolitan Institute of Medical Science, Tokyo, Japan
- AMED-CREST, Japan Agency for Medical Research and Development, Tokyo, Japan
- Correspondence should be addressed: M. Murakami, Laboratory of Environmental and Metabolic Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, the University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8655, Japan (e-mail: )
| |
Collapse
|
42
|
Roos L, Sandling JK, Bell CG, Glass D, Mangino M, Spector TD, Deloukas P, Bataille V, Bell JT. Higher Nevus Count Exhibits a Distinct DNA Methylation Signature in Healthy Human Skin: Implications for Melanoma. J Invest Dermatol 2016; 137:910-920. [PMID: 27993549 PMCID: PMC5754330 DOI: 10.1016/j.jid.2016.11.029] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 10/14/2016] [Accepted: 11/21/2016] [Indexed: 12/13/2022]
Abstract
High nevus count is the strongest risk factor for melanoma, and although gene variants have been discovered for both traits, epigenetic variation is unexplored. We investigated 322 healthy human skin DNA methylomes associated with total body nevi count, incorporating genetic and transcriptomic variation. DNA methylation changes were identified at genes involved in melanocyte biology, such as RAF1 (P = 1.2 × 10-6) and CTC1 (region: P = 6.3 × 10-4), and other genes including ARRDC1 (P = 3.1 × 10-7). A subset exhibited coordinated methylation and transcription changes within the same biopsy. The total analysis was also enriched for melanoma-associated DNA methylation variation (P = 6.33 × 10-6). In addition, we show that skin DNA methylation is associated in cis with known genome-wide association study single nucleotide polymorphisms for nevus count, at PLA2G6 (P = 1.7 × 10-49) and NID1 (P = 6.4 × 10-14), as well as melanoma risk, including in or near MC1R, MX2, and TERT/CLPTM1L (P < 1 × 10-10). Our analysis using a uniquely large dataset comprising healthy skin DNA methylomes identified known and additional regulatory loci and pathways in nevi and melanoma biology. This integrative study improves our understanding of predisposition to nevi and their potential contribution to melanoma pathogenesis.
Collapse
Affiliation(s)
- Leonie Roos
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK; MRC London Institute of Medical Sciences, London, UK; Institute of Clinical Sciences, Faculty of Medicine, Imperial College London, London, UK.
| | - Johanna K Sandling
- Department of Medical Sciences, Molecular Medicine and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Christopher G Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK; MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK; Human Development and Health Academic Unit, Institute of Developmental Sciences, University of Southampton, Southampton, UK; Epigenomic Medicine, Centre for Biological Sciences, Faculty of Environmental and Natural Sciences, University of Southampton, Southampton, UK
| | - Daniel Glass
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Tim D Spector
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Panos Deloukas
- William Harvey Research Institute, Queen Mary University of London, London, UK
| | - Veronique Bataille
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| | - Jordana T Bell
- Department of Twin Research and Genetic Epidemiology, King's College London, London, UK
| |
Collapse
|
43
|
Zhi L, Liu D, Wu SG, Li T, Zhao G, Zhao B, Li M. Association of common variants in MTAP with susceptibility and overall survival of osteosarcoma: a two-stage population-based study in Han Chinese. J Cancer 2016; 7:2179-2186. [PMID: 27994653 PMCID: PMC5166526 DOI: 10.7150/jca.16609] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2016] [Accepted: 09/04/2016] [Indexed: 12/19/2022] Open
Abstract
Osteosarcoma (OS) is a common malignant tumor, which exists widely in the bone of children and adolescents, and genetic factors may influence its susceptibility. Recently, the gene MTAP has been reported to be associated with OS in a Caucasian population. To investigate the association of common variants in MTAP with OS risk in Han Chinese individuals, we designed a two-stage case-control study with 392 OS patients and 1,578 unrelated healthy controls of Han Chinese individuals. A total of 17 tagging single nucleotide polymorphisms (SNPs) were firstly genotyped in the discovery stage, and single-SNP association and haplotypic association analyses have been performed. The SNP rs7023329 was found to be strongly associated with the OS risk (adjusted P = 0.002908), and the results of odds ratios (ORs) and 95% confidence intervals (CI) revealed increased risks from A allele of the SNP on OS (OR=1.33, 95% CI=1.13-1.62). The results were confirmed with a similar pattern in the validation stage (adjusted P = 0.006737, OR=1.49, 95% CI=1.11-2.00). Moreover, haplotypic analyses indicated that one haplotype block containing rs7023329 was significantly associated with OS risk in both stages (both global P<0.0001). The statistically significant association between the rs7023329 genotype and poor survival in OS patients was also observed. To sum up, our results prove that MTAP plays an important role in the etiology of OS, suggesting this gene as a potential genetic modifier for OS development.
Collapse
Affiliation(s)
- Liqiang Zhi
- Department of Orthopedic, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Dan Liu
- Department of Rheumatology and Immunology, the Fifth Hospital of Xi'an, Xi'an, China
| | - Stephen G Wu
- Department of Energy, Environment and Chemical Engineering, Washington University, Saint Louis, MO, USA
| | - Tianqing Li
- Department of Orthopedic, Xijing Orthopedic Hospital, the Fourth Military Medical University, Xi'an, China
| | - Guanghui Zhao
- Department of Orthopedic, Hong-hui Hospital, Xi'an Jiaotong University College of Medicine, Xi'an, China
| | - Bo Zhao
- Department of Orthopedic, the Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Meng Li
- Department of Orthopedic, the First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
44
|
Taylor NJ, Thomas NE, Anton-Culver H, Armstrong BK, Begg CB, Busam KJ, Cust AE, Dwyer T, From L, Gallagher RP, Gruber SB, Nishri DE, Orlow I, Rosso S, Venn AJ, Zanetti R, Berwick M, Kanetsky PA. Nevus count associations with pigmentary phenotype, histopathological melanoma characteristics and survival from melanoma. Int J Cancer 2016; 139:1217-22. [PMID: 27101944 PMCID: PMC4939099 DOI: 10.1002/ijc.30157] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 03/25/2016] [Accepted: 04/06/2016] [Indexed: 12/31/2022]
Abstract
Although nevus count is an established risk factor for melanoma, relationships between nevus number and patient and tumor characteristics have not been well studied and the influence of nevus count on melanoma-specific survival is equivocal. Using data from the Genes, Environment and Melanoma (GEM) study, a large population-based study of primary cutaneous melanoma, we evaluated associations between number of nevi and patient features, including sun-sensitivity summarized in a phenotypic index, and tumor characteristics. We also assessed the association of nevus count with melanoma-specific survival. Higher nevus counts were independently and positively associated with male gender and younger age at diagnosis, and they were inversely associated with lentigo maligna histology. We observed a borderline significant trend of poorer melanoma-specific survival with increasing quartile of nevus count, but little or no association between number of nevi and pigmentary phenotypic characteristics or prognostic tumor features.
Collapse
Affiliation(s)
- Nicholas J. Taylor
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Nancy E. Thomas
- Department of Dermatology, Lineberger Comprehensive Cancer Center, University of North Carolina, Chapel Hill, NC, USA
| | - Hoda Anton-Culver
- Department of Epidemiology, University of California, Irvine, CA, USA
| | | | - Colin B. Begg
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Klaus J. Busam
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anne E. Cust
- School of Public Health, University of Sydney, Sydney, NSW, Australia
| | - Terence Dwyer
- George Institute for Global Health Research, University of Oxford, 34 Broad St. OX1 3BD, Oxford, UK
| | - Lynn From
- Women’s College Hospital, Toronto, ON, Canada
| | | | - Stephen B. Gruber
- Keck School of Medicine, University of Southern California Norris Comprehensive Cancer Center, Los Angeles, CA, USA
| | | | - Irene Orlow
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | - Alison J. Venn
- Menzies Centre for Population Health, University of Tasmania, Hobart, Tasmania, Australia
| | | | - Marianne Berwick
- Departments of Internal Medicine and Dermatology, University of New Mexico, Albuquerque, NM, USA
| | - Peter A. Kanetsky
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | |
Collapse
|
45
|
Dechamethakun S, Muramatsu M. Long noncoding RNA variations in cardiometabolic diseases. J Hum Genet 2016; 62:97-104. [PMID: 27305986 DOI: 10.1038/jhg.2016.70] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2016] [Revised: 04/27/2016] [Accepted: 05/06/2016] [Indexed: 12/18/2022]
Abstract
Cardiometabolic diseases are characterized as a combination of multiple risk factors for cardiovascular disease (CVD) and metabolic diseases including diabetes mellitus, dyslipidemia, hypertension and abdominal obesity. This cluster of abnormalities individually and interdependently leads to atherosclerosis and CVD morbidity and mortality. In the past decade, genome-wide association studies (GWASs) have identified a series of cardiometabolic disease-associated variants that can collectively explain a small proportion of the variability. Intriguingly, the susceptibility variants imputed from GWASs usually do not reside in the coding regions, suggesting a crucial role of the noncoding elements of the genome. In recent years, emerging evidence suggests that noncoding RNA (ncRNA) is functional for physiology and pathophysiology of human diseases. These include microRNAs and long noncoding RNAs (lncRNAs) that are now implicated in human diseases. The ncRNAs can interact with each other and with proteins, to interfere gene expressions, leading to the development of many human disorders. Although evidence suggests the functional role of lncRNAs in cardiometabolic traits, the molecular mechanisms of gene regulation underlying cardiometabolic diseases remain to be better defined. Here, we summarize the recent discoveries of lncRNA variations in the context of cardiometabolic diseases.
Collapse
Affiliation(s)
- Sariya Dechamethakun
- Department of Molecular Epidemiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | - Masaaki Muramatsu
- Department of Molecular Epidemiology, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
46
|
Zhang T, Dutton-Regester K, Brown KM, Hayward NK. The genomic landscape of cutaneous melanoma. Pigment Cell Melanoma Res 2016; 29:266-83. [PMID: 26833684 DOI: 10.1111/pcmr.12459] [Citation(s) in RCA: 127] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 01/25/2016] [Indexed: 12/31/2022]
Abstract
Somatic mutation analysis of melanoma has been performed at the single gene level extensively over the past several decades. This has provided considerable insight into the critical pathways controlling melanoma initiation and progression. During the last 5 yr, next-generation sequencing (NGS) has enabled even more comprehensive mutational screening at the level of multigene panels, exomes and genomes. These studies have uncovered many new and unexpected players in melanoma development. The recent landmark study from The Cancer Genome Atlas (TCGA) consortium describing the genomic architecture of 333 cutaneous melanomas provides the largest and broadest analysis to date on the somatic aberrations underlying melanoma genesis. It thus seems timely to review the mutational landscape of melanoma and highlight the key genes and cellular pathways that appear to drive this cancer.
Collapse
Affiliation(s)
- Tongwu Zhang
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Ken Dutton-Regester
- Cancer Program, Broad Institute of Harvard and MIT, Cambridge, MA, USA
- Department of Medical Oncology, Dana Farber Cancer Institute, Boston, MA, USA
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| | - Kevin M Brown
- Laboratory of Translational Genomics, Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Nicholas K Hayward
- Oncogenomics Laboratory, QIMR Berghofer Medical Research Institute, Herston, Qld, Australia
| |
Collapse
|
47
|
The role of the melanoma gene MC1R in Parkinson disease and REM sleep behavior disorder. Neurobiol Aging 2016; 43:180.e7-180.e13. [PMID: 27131830 DOI: 10.1016/j.neurobiolaging.2016.03.029] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2016] [Revised: 03/20/2016] [Accepted: 03/28/2016] [Indexed: 01/16/2023]
Abstract
The MC1R gene, suggested to be involved in Parkinson disease (PD) and melanoma, was sequenced in PD patients (n = 539) and controls (n = 265) from New York, and PD patients (n = 551), rapid eye movement sleep behavior disorder (RBD) patients (n = 351), and controls (n = 956) of European ancestry. Sixty-eight MC1R variants were identified, including 7 common variants with frequency > 0.01. None of the common variants was associated with PD or RBD in the different regression models. In a meta-analysis with fixed-effect model, the p.R160W variant was associated with an increased risk for PD (odds ratio = 1.22, 95% confidence interval = 1.02-1.47, p = 0.03) but with significant heterogeneity (p = 0.048). Removing one study that introduced the heterogeneity resulted in nonsignificant association (odds ratio = 1.11, 95% confidence interval, 0.92-1.35, p = 0.27, heterogeneity p = 0.57). Rare variants had similar frequencies in patients and controls (10.54% and 10.15%, respectively, p = 0.75), and no cumulative effect of carrying more than one MC1R variant was found. The present study does not support a role for the MC1R p.R160W and other variants in susceptibility for PD or RBD.
Collapse
|
48
|
Johansen P, Andersen JD, Madsen LN, Ullum H, Glud M, Børsting C, Gniadecki R, Morling N. Pigmentary Markers in Danes--Associations with Quantitative Skin Colour, Nevi Count, Familial Atypical Multiple-Mole, and Melanoma Syndrome. PLoS One 2016; 11:e0150381. [PMID: 26938746 PMCID: PMC4777533 DOI: 10.1371/journal.pone.0150381] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Accepted: 02/12/2016] [Indexed: 12/15/2022] Open
Abstract
To investigate whether pigmentation genes involved in the melanogenic pathway (melanogenesis) contributed to melanoma predisposition, we compared pigmentary genetics with quantitative skin pigmentation measurements, the number of atypical nevi, the total nevus count, and the familial atypical multiple mole and melanoma (FAMMM) syndrome. We typed 32 pigmentary SNP markers and sequenced MC1R in 246 healthy individuals and 116 individuals attending periodic control for malignant melanoma development, 50 of which were diagnosed with FAMMM. It was observed that individuals with any two grouped MC1R variants (missense, NM_002386:c. 456C > A (p.TYR152*), or NM_002386:c.83_84insA (p.Asn29Glnfs*14) had significantly (p<0.001) lighter skin pigmentation of the upper-inner arm than those with none or one MC1R variant. We did not observe any significant association of the MC1R variants with constitutive pigmentation measured on the buttock area. We hypothesize that the effect of MC1R variants on arm pigmentation is primarily reflecting the inability to tan when subjected to UVR. A gender specific effect on skin pigmentation was also observed, and it was found that the skin pigmentation of females on average were darker than that of males (p<0.01). We conclude that MC1R variants are associated with quantitative skin colour in a lightly pigmented Danish population. We did not observe any association between any pigmentary marker and the FAMMM syndrome. We suggest that the genetics of FAMMM is not related to the genetics of the pigmentary pathway.
Collapse
Affiliation(s)
- Peter Johansen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Jeppe Dyrberg Andersen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Linnea Nørgård Madsen
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Henrik Ullum
- Department of Clinical Immunology, University Hospital Copenhagen, Rigshospitalet, Copenhagen, Denmark
| | - Martin Glud
- Department of Dermatology, Faculty of Health and Medical Sciences, University of Copenhagen, Bispebjerg Hospital, Copenhagen, Denmark
| | - Claus Børsting
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Robert Gniadecki
- Department of Dermatology, Faculty of Health and Medical Sciences, University of Copenhagen, Bispebjerg Hospital, Copenhagen, Denmark
| | - Niels Morling
- Section of Forensic Genetics, Department of Forensic Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
- * E-mail:
| |
Collapse
|
49
|
Lu Y, Day FR, Gustafsson S, Buchkovich ML, Na J, Bataille V, Cousminer DL, Dastani Z, Drong AW, Esko T, Evans DM, Falchi M, Feitosa MF, Ferreira T, Hedman ÅK, Haring R, Hysi PG, Iles MM, Justice AE, Kanoni S, Lagou V, Li R, Li X, Locke A, Lu C, Mägi R, Perry JRB, Pers TH, Qi Q, Sanna M, Schmidt EM, Scott WR, Shungin D, Teumer A, Vinkhuyzen AAE, Walker RW, Westra HJ, Zhang M, Zhang W, Zhao JH, Zhu Z, Afzal U, Ahluwalia TS, Bakker SJL, Bellis C, Bonnefond A, Borodulin K, Buchman AS, Cederholm T, Choh AC, Choi HJ, Curran JE, de Groot LCPGM, De Jager PL, Dhonukshe-Rutten RAM, Enneman AW, Eury E, Evans DS, Forsen T, Friedrich N, Fumeron F, Garcia ME, Gärtner S, Han BG, Havulinna AS, Hayward C, Hernandez D, Hillege H, Ittermann T, Kent JW, Kolcic I, Laatikainen T, Lahti J, Leach IM, Lee CG, Lee JY, Liu T, Liu Y, Lobbens S, Loh M, Lyytikäinen LP, Medina-Gomez C, Michaëlsson K, Nalls MA, Nielson CM, Oozageer L, Pascoe L, Paternoster L, Polašek O, Ripatti S, Sarzynski MA, Shin CS, Narančić NS, Spira D, Srikanth P, Steinhagen-Thiessen E, Sung YJ, Swart KMA, Taittonen L, Tanaka T, et alLu Y, Day FR, Gustafsson S, Buchkovich ML, Na J, Bataille V, Cousminer DL, Dastani Z, Drong AW, Esko T, Evans DM, Falchi M, Feitosa MF, Ferreira T, Hedman ÅK, Haring R, Hysi PG, Iles MM, Justice AE, Kanoni S, Lagou V, Li R, Li X, Locke A, Lu C, Mägi R, Perry JRB, Pers TH, Qi Q, Sanna M, Schmidt EM, Scott WR, Shungin D, Teumer A, Vinkhuyzen AAE, Walker RW, Westra HJ, Zhang M, Zhang W, Zhao JH, Zhu Z, Afzal U, Ahluwalia TS, Bakker SJL, Bellis C, Bonnefond A, Borodulin K, Buchman AS, Cederholm T, Choh AC, Choi HJ, Curran JE, de Groot LCPGM, De Jager PL, Dhonukshe-Rutten RAM, Enneman AW, Eury E, Evans DS, Forsen T, Friedrich N, Fumeron F, Garcia ME, Gärtner S, Han BG, Havulinna AS, Hayward C, Hernandez D, Hillege H, Ittermann T, Kent JW, Kolcic I, Laatikainen T, Lahti J, Leach IM, Lee CG, Lee JY, Liu T, Liu Y, Lobbens S, Loh M, Lyytikäinen LP, Medina-Gomez C, Michaëlsson K, Nalls MA, Nielson CM, Oozageer L, Pascoe L, Paternoster L, Polašek O, Ripatti S, Sarzynski MA, Shin CS, Narančić NS, Spira D, Srikanth P, Steinhagen-Thiessen E, Sung YJ, Swart KMA, Taittonen L, Tanaka T, Tikkanen E, van der Velde N, van Schoor NM, Verweij N, Wright AF, Yu L, Zmuda JM, Eklund N, Forrester T, Grarup N, Jackson AU, Kristiansson K, Kuulasmaa T, Kuusisto J, Lichtner P, Luan J, Mahajan A, Männistö S, Palmer CD, Ried JS, Scott RA, Stancáková A, Wagner PJ, Demirkan A, Döring A, Gudnason V, Kiel DP, Kühnel B, Mangino M, Mcknight B, Menni C, O'Connell JR, Oostra BA, Shuldiner AR, Song K, Vandenput L, van Duijn CM, Vollenweider P, White CC, Boehnke M, Boettcher Y, Cooper RS, Forouhi NG, Gieger C, Grallert H, Hingorani A, Jørgensen T, Jousilahti P, Kivimaki M, Kumari M, Laakso M, Langenberg C, Linneberg A, Luke A, Mckenzie CA, Palotie A, Pedersen O, Peters A, Strauch K, Tayo BO, Wareham NJ, Bennett DA, Bertram L, Blangero J, Blüher M, Bouchard C, Campbell H, Cho NH, Cummings SR, Czerwinski SA, Demuth I, Eckardt R, Eriksson JG, Ferrucci L, Franco OH, Froguel P, Gansevoort RT, Hansen T, Harris TB, Hastie N, Heliövaara M, Hofman A, Jordan JM, Jula A, Kähönen M, Kajantie E, Knekt PB, Koskinen S, Kovacs P, Lehtimäki T, Lind L, Liu Y, Orwoll ES, Osmond C, Perola M, Pérusse L, Raitakari OT, Rankinen T, Rao DC, Rice TK, Rivadeneira F, Rudan I, Salomaa V, Sørensen TIA, Stumvoll M, Tönjes A, Towne B, Tranah GJ, Tremblay A, Uitterlinden AG, van der Harst P, Vartiainen E, Viikari JS, Vitart V, Vohl MC, Völzke H, Walker M, Wallaschofski H, Wild S, Wilson JF, Yengo L, Bishop DT, Borecki IB, Chambers JC, Cupples LA, Dehghan A, Deloukas P, Fatemifar G, Fox C, Furey TS, Franke L, Han J, Hunter DJ, Karjalainen J, Karpe F, Kaplan RC, Kooner JS, McCarthy MI, Murabito JM, Morris AP, Bishop JAN, North KE, Ohlsson C, Ong KK, Prokopenko I, Richards JB, Schadt EE, Spector TD, Widén E, Willer CJ, Yang J, Ingelsson E, Mohlke KL, Hirschhorn JN, Pospisilik JA, Zillikens MC, Lindgren C, Kilpeläinen TO, Loos RJF. New loci for body fat percentage reveal link between adiposity and cardiometabolic disease risk. Nat Commun 2016; 7:10495. [PMID: 26833246 PMCID: PMC4740398 DOI: 10.1038/ncomms10495] [Show More Authors] [Citation(s) in RCA: 200] [Impact Index Per Article: 22.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Accepted: 12/16/2015] [Indexed: 12/24/2022] Open
Abstract
To increase our understanding of the genetic basis of adiposity and its links to cardiometabolic disease risk, we conducted a genome-wide association meta-analysis of body fat percentage (BF%) in up to 100,716 individuals. Twelve loci reached genome-wide significance (P<5 × 10(-8)), of which eight were previously associated with increased overall adiposity (BMI, BF%) and four (in or near COBLL1/GRB14, IGF2BP1, PLA2G6, CRTC1) were novel associations with BF%. Seven loci showed a larger effect on BF% than on BMI, suggestive of a primary association with adiposity, while five loci showed larger effects on BMI than on BF%, suggesting association with both fat and lean mass. In particular, the loci more strongly associated with BF% showed distinct cross-phenotype association signatures with a range of cardiometabolic traits revealing new insights in the link between adiposity and disease risk.
Collapse
Affiliation(s)
- Yingchang Lu
- The Charles Bronfman Institute for Personalized Medicine, The
Icahn School of Medicine at Mount Sinai, New York, New
York
10029, USA
- The Department of Preventive Medicine, The Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
| | - Felix R. Day
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Stefan Gustafsson
- Science for Life Laboratory, Uppsala University, 750
85
Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, 751 85
Uppsala, Sweden
| | - Martin L. Buchkovich
- Department of Genetics, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
| | - Jianbo Na
- Department of Developmental and Regenerative Biology, The Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| | - Veronique Bataille
- West Herts NHS Trust, Herts
HP2 4AD, UK
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Diana L. Cousminer
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
| | - Zari Dastani
- Department Epidemiology, Biostatistics and Human Genetics, Lady
Davis Institute, Jewish General Hospital, McGill University,
Montréal, Quebec, Canada
H3T1E2
| | - Alexander W. Drong
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Tõnu Esko
- Estonian Genome Center, Univeristy of Tartu,
Tartu, 51010, Estonia
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
| | - David M. Evans
- University of Queensland Diamantina Institute, Translational
Research Institute, Brisbane, Queensland
4102, Australia
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
| | - Mario Falchi
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- Department of Genomics of Common Disease, School of Public
Health, Imperial College London, London
W12 0NN, UK
| | - Mary F. Feitosa
- Division of Statistical Genomics, Department of Genetics,
Washington University School of Medicine, St Louis,
Missouri
63108, USA
| | - Teresa Ferreira
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Åsa K. Hedman
- Science for Life Laboratory, Uppsala University, 750
85
Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, 751 85
Uppsala, Sweden
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Robin Haring
- Institute of Clinical Chemistry and Laboratory Medicine,
University Medicine Greifswald, 17475
Greifswald, Germany
- European University of Applied Sciences, Faculty of Applied
Public Health, 18055
Rostock, Germany
| | - Pirro G. Hysi
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Mark M. Iles
- Leeds Institute of Cancer and Pathology, Cancer Research UK
Leeds Centre, University of Leeds, Leeds
LS9 7TF, UK
| | - Anne E. Justice
- Department of Epidemiology, University of North Carolina at
Chapel Hill, Chapel Hill, North Carolina
27599, USA
| | - Stavroula Kanoni
- William Harvey Research Institute, Barts and The London School
of Medicine and Dentistry, Queen Mary University of London,
London
EC1M 6BQ, UK
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
| | - Vasiliki Lagou
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
| | - Rui Li
- Department Epidemiology, Biostatistics and Human Genetics, Lady
Davis Institute, Jewish General Hospital, McGill University,
Montréal, Quebec, Canada
H3T1E2
| | - Xin Li
- Department of Epidemiology, Harvard School of Public
Health, Boston, Massachusetts
02115, USA
| | - Adam Locke
- Center for Statistical Genetics, Department of Biostatistics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Chen Lu
- Department of Biostatistics, Boston University School of Public
Health, Boston, Massachusetts
02118, USA
| | - Reedik Mägi
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Estonian Genome Center, Univeristy of Tartu,
Tartu, 51010, Estonia
| | - John R. B. Perry
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Tune H. Pers
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Medical and Population Genetics Program, Broad Institute of MIT
and Harvard, Cambridge
02142, USA
- Department of Epidemiology Research, Statens Serum
Institut, 2100
Copenhagen, Denmark
| | - Qibin Qi
- Department of Epidemiology and Popualtion Health, Albert
Einstein College of Medicine, Bronx, New York
10461, USA
| | - Marianna Sanna
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
- Department of Genomics of Common Disease, School of Public
Health, Imperial College London, London
W12 0NN, UK
| | - Ellen M. Schmidt
- Department of Computational Medicine and Bioinformatics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - William R. Scott
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
| | - Dmitry Shungin
- Lund University Diabetes Centre, Department of Clinical
Science, Genetic and Molecular Epidemiology Unit, Skåne University
Hosptial, 205 02
Malmö, Sweden
- Department of Public Health and Clinical Medicine, Unit of
Medicine, Umeå University, 901 87
Umeå, Sweden
- Department of Odontology, Umeå University,
901 85
Umeå, Sweden
| | - Alexander Teumer
- Institute for Community Medicine, University Medicine
Greifswald, 17475
Greifswald, Germany
- Interfaculty Institute for Genetics and Functional Genomics,
University Medicine Greifswald, 17475
Greifswald, Germany
| | | | - Ryan W. Walker
- The Charles Bronfman Institute for Personalized Medicine, The
Icahn School of Medicine at Mount Sinai, New York, New
York
10029, USA
- The Department of Preventive Medicine, The Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
| | - Harm-Jan Westra
- Program in Medical and Population Genetics, Broad Institute of
Harvard and Massachusetts Institute of Technology, Cambridge,
Massachusetts
02142, USA
- Divisions of Genetics and Rheumatology, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachusetts
02446, USA
- Partners Center for Personalized Genetic Medicine,
Boston, Massachusetts
02446, USA
| | - Mingfeng Zhang
- Department of Dermatology, Brigham and Women's
Hospital, Boston, Massachusetts
02115, USA
| | - Weihua Zhang
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
| | - Jing Hua Zhao
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Zhihong Zhu
- Queensland Brain Institute, The University of Queensland,
Brisbane
4072, Australia
| | - Uzma Afzal
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
| | - Tarunveer Singh Ahluwalia
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Copenhagen Prospective Studies on Asthma in Childhood, Faculty
of Health and Medical Sceinces, University of Copenhagen, 2200
Copenhagen, Denmark
- Danish Pediatric Asthma Center, Gentofte Hospital, The Capital
Region, 2200
Copenhagen, Denmark
- Steno Diabetes Center A/S, DK-2820
Gentofte, Denmark
| | - Stephan J. L. Bakker
- University of Groningen, University Medical Center Groningen,
Department of Medicine, 9700 RB
Groningen, The Netherlands
| | - Claire Bellis
- Department of Genetics, Texas Biomedical Research
Institute, San Antonio, Texas
78245, USA
| | - Amélie Bonnefond
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Katja Borodulin
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Aron S. Buchman
- Rush Alzheimer's Disease Center, Rush University
Medical Center, Chicago, Illinois
60612, USA
| | - Tommy Cederholm
- Department of Public Health and Caring Sciences, Clinical
Nutrition and Metabolism, Uppsala University, 751 85
Uppsala, Sweden
| | - Audrey C. Choh
- Lifespan Health Research Center, Wright State University
Boonshoft School of Medicine, Dayton, Ohio
45420, USA
| | - Hyung Jin Choi
- Department of Anatomy, Seoul National University College of
Medicine, Seoul
03080, Korea
| | - Joanne E. Curran
- South Texas Diabetes and Obesity Institute, University of Texas
Rio Grande Valley, Brownsville, Texas
78520
| | | | - Philip L. De Jager
- Program in Medical and Population Genetics, Broad Institute of
Harvard and Massachusetts Institute of Technology, Cambridge,
Massachusetts
02142, USA
- Harvard Medical School, Boston,
Massachusetts
02115, USA
- Program in Translational NeuroPsychiatric Genomics, Department
of Neurology, Brigham and Women's Hospital, Boston,
Massachusetts
02115, USA
| | | | - Anke W. Enneman
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Elodie Eury
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Daniel S. Evans
- California Pacific Medical Center Research Institute,
San Francisco, California
94107, USA
| | - Tom Forsen
- Department of General Practice and Primary Health Care,
University of Helsinki, FI-00014
Helsinki, Finland
| | - Nele Friedrich
- Institute of Clinical Chemistry and Laboratory Medicine,
University Medicine Greifswald, 17475
Greifswald, Germany
| | - Frédéric Fumeron
- INSERM, UMR_S 1138, Centre de Recherche des Cordeliers,
F-75006
Paris, France
- Sorbonne Universités, UPMC Univ Paris 06, UMR_S
1138, Centre de Recherche des Cordeliers, F-75006
Paris, France
- Université Paris Descartes, Sorbonne Paris
Cité, UMR_S 1138, Centre de Recherche des Cordeliers,
F-75006
Paris, France
- Univ Paris Diderot, Sorbonne Paris Cité, UMR_S 1138,
Centre de Recherche des Cordeliers, F-75006
Paris, France
| | - Melissa E. Garcia
- Laboratory of Epidemiology and Population Sciences, National
Institute on Aging, Bethesda, Maryland
20892, USA
| | - Simone Gärtner
- Department of Medicine A, University Medicine Greifswald,
17475
Greifswald, Germany
| | - Bok-Ghee Han
- Center for Genome Science, National Institute of Health, Osong
Health Technology Administration Complex, Chungcheongbuk-do
370914, Korea
| | - Aki S. Havulinna
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Caroline Hayward
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Dena Hernandez
- Laboratory of Neurogenetics, National Institute on Aging,
National Institutes of Health, Bethesda, Maryland
20892, USA
| | - Hans Hillege
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
| | - Till Ittermann
- Institute for Community Medicine, University Medicine
Greifswald, 17475
Greifswald, Germany
| | - Jack W. Kent
- Department of Genetics, Texas Biomedical Research
Institute, San Antonio, Texas
78245, USA
| | - Ivana Kolcic
- Department of Public Health, Faculty of Medicine, University of
Split, Split
21000, Croatia
| | - Tiina Laatikainen
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
- Hospital District of North Karelia, FI-80210
Joensuu, Finland
- Institute of Public Health and Clinical Nutrition, University
of Eastern Finland, FI-70211
Kuopio, Finland
| | - Jari Lahti
- Folkhälsan Research Centre, FI-00290
Helsinki, Finland
- Institute of Behavioural Sciences, University of
Helsinki, FI-00014
Helsinki, Finland
| | - Irene Mateo Leach
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
| | - Christine G. Lee
- Department of Medicine, Oregon Health and Science
University, Portland, Oregon
97239, USA
- Research Service, Veterans Affairs Medical Center,
Portland, Oregon
97239, USA
| | - Jong-Young Lee
- Center for Genome Science, National Institute of Health, Osong
Health Technology Administration Complex, Chungcheongbuk-do
370914, Korea
| | - Tian Liu
- Max Planck Institute for Molecular Genetics, Department of
Vertebrate Genomics, 14195
Berlin, Germany
- Max Planck Institute for Human Development,
14194
Berlin, Germany
| | - Youfang Liu
- Thurston Arthritis Research Center, University of North
Carolina at Chapel Hill, Chaper Hill, North Carolina
27599-7280, USA
| | - Stéphane Lobbens
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Marie Loh
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Translational Laboratory in Genetic Medicine (TLGM), Agency for
Science, Technology and Research (A*STAR), 8A Biomedical
Grove, Immunos, Level 5, Singapore
138648, Singapore
| | - Leo-Pekka Lyytikäinen
- Department of Clinical Chemistry, University of Tampere School
of Medicine, FI-33014
Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and
School of Medicine, University of Tampere, FI-33520
Tampere, Finland
| | - Carolina Medina-Gomez
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Karl Michaëlsson
- Department of Surgical Sciences, Orthopedics, Uppsala
University, 751 85
Uppsala, Sweden
| | - Mike A. Nalls
- Laboratory of Neurogenetics, National Institute on Aging,
National Institutes of Health, Bethesda, Maryland
20892, USA
| | - Carrie M. Nielson
- School of Public Health, Oregon Health & Science
University, Portland, Oregon
97239, USA
- Bone & Mineral Unit, Oregon Health & Science
University, Portland, Oregon
97239, USA
| | | | - Laura Pascoe
- Institute of Cell & Molecular Biosciences, Newcastle
University, Newcastle
NE1 7RU, UK
| | - Lavinia Paternoster
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
| | - Ozren Polašek
- Department of Public Health, Faculty of Medicine, University of
Split, Split
21000, Croatia
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Samuli Ripatti
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
- Hjelt Institute, University of Helsinki,
FI-00014
Helsinki, Finland
| | - Mark A. Sarzynski
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - Chan Soo Shin
- Department of Internal Medicine, Seoul National University
College of Medicine, Seoul
03080, Korea
| | | | - Dominik Spira
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
- Lipid Clinic at the Interdisciplinary Metabolism Center,
Charité-Universitätsmedizin Berlin, 13353
Berlin, Germany
| | - Priya Srikanth
- School of Public Health, Oregon Health & Science
University, Portland, Oregon
97239, USA
- Bone & Mineral Unit, Oregon Health & Science
University, Portland, Oregon
97239, USA
| | - Elisabeth Steinhagen-Thiessen
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
- Lipid Clinic at the Interdisciplinary Metabolism Center,
Charité-Universitätsmedizin Berlin, 13353
Berlin, Germany
| | - Yun Ju Sung
- Division of Biostatistics, Washington University School of
Medicine, St Louis, Missouri
63110, USA
| | - Karin M. A. Swart
- EMGO Institute for Health and Care Research, VU University
Medical Center, 1081 BT
Amsterdam, The Netherlands
- VUMC, Department of Epidemiology and Biostatistics,
1081 BT
Amsterdam, The Netherlands
| | - Leena Taittonen
- Department of Pediatrics, University of Oulu,
FI-90014
Oulu, Finland
- Department of Pediatrics, Vaasa Central Hospital,
FI-65100
Vaasa, Finland
| | - Toshiko Tanaka
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Emmi Tikkanen
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Hjelt Institute, University of Helsinki,
FI-00014
Helsinki, Finland
| | - Nathalie van der Velde
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Natasja M. van Schoor
- EMGO Institute for Health and Care Research, VU University
Medical Center, 1081 BT
Amsterdam, The Netherlands
- VUMC, Department of Epidemiology and Biostatistics,
1081 BT
Amsterdam, The Netherlands
| | - Niek Verweij
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
| | - Alan F. Wright
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Lei Yu
- Rush Alzheimer's Disease Center, Rush University
Medical Center, Chicago, Illinois
60612, USA
| | - Joseph M. Zmuda
- Department of Epidemiology; University of Pittsburgh,
Pittsburgh, Pennsylvania
15261, USA
| | - Niina Eklund
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Terrence Forrester
- Tropical Metabolism Research Unit, Tropical Medicine Research
Institute, University of the West Indies, Mona
JMAAW15, Jamaica
| | - Niels Grarup
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
| | - Anne U. Jackson
- Center for Statistical Genetics, Department of Biostatistics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Kati Kristiansson
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Teemu Kuulasmaa
- Faculty of Health Sciences, Institute of Clinical Medicine,
Internal Medicine, University of Eastern Finland, 70210
Kuopio, Finland
| | - Johanna Kuusisto
- Faculty of Health Sciences, Institute of Clinical Medicine,
Internal Medicine, University of Eastern Finland, 70210
Kuopio, Finland
- Department of Medicine, University of Eastern Finland,
70210
Kuopio, Finland
- Kuopio University Hospital, 70029
Kuopio, Finland
| | - Peter Lichtner
- Institute of Human Genetics, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Jian'an Luan
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Anubha Mahajan
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
| | - Satu Männistö
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Cameron D. Palmer
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
| | - Janina S. Ried
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Robert A. Scott
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Alena Stancáková
- Department of Medicine, University of Eastern Finland and
Kuopio University Hospital, 70210
Kuopio, Finland
| | - Peter J. Wagner
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Ayse Demirkan
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
University Medical Center, 3015GE
Rotterdam, The Netherlands
| | - Angela Döring
- Institute of Epidemiology I, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Vilmundur Gudnason
- Icelandic Heart Association, Kopavogur
201, Iceland
- University of Iceland, Faculty of Medicine,
Reykjavik
101, Iceland
| | - Douglas P. Kiel
- Department of Medicine Beth Israel Deaconess Medical Center
and Harvard Medical School, Boston, Massachusetts
02115
- Institute for Aging Research Hebrew Senior Life,
Boston, Massachusetts
02131, USA
| | - Brigitte Kühnel
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Massimo Mangino
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Barbara Mcknight
- Cardiovascular Health Research Unit, University of
Washington, Seattle, Washington
98101, USA
- Program in Biostatistics and Biomathematics, Divison of Public
Health Sciences, Fred Hutchinson Cancer Research Center,
Seattle, Washington
98109, USA
- Department of Biostatistics, University of Washington,
Seattle, Washington
98195, USA
| | - Cristina Menni
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Jeffrey R. O'Connell
- Program for Personalized and Genomic Medicine, Division of
Endocrinology, Diabetes and Nutrition, Department of Medicine, University of
Maryland School of Medicine, Baltimore, Maryland
21201, USA
| | - Ben A. Oostra
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
University Medical Center, 3015GE
Rotterdam, The Netherlands
| | - Alan R. Shuldiner
- Program for Personalized and Genomic Medicine, Division of
Endocrinology, Diabetes and Nutrition, Department of Medicine, University of
Maryland School of Medicine, Baltimore, Maryland
21201, USA
- Geriatric Research and Education Clinical Center, Vetrans
Administration Medical Center, Baltimore, Maryland
21042, USA
| | - Kijoung Song
- Genetics, Projects Clinical Platforms and Sciences,
GlaxoSmithKline, Philadelphia, Pennsylvania
19112, USA
| | - Liesbeth Vandenput
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, 413 45
Gothenburg, Sweden
| | - Cornelia M. van Duijn
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Genetic Epidemiology Unit, Department of Epidemiology, Erasmus
University Medical Center, 3015GE
Rotterdam, The Netherlands
- Center for Medical Systems Biology, 2300
Leiden, The Netherlands
| | - Peter Vollenweider
- Department of Internal Medicine, University Hospital Lausanne
(CHUV) and University of Lausanne, 1011
Lausanne, Switzerland
| | - Charles C. White
- Department of Biostatistics, Boston University School of Public
Health, Boston, Massachusetts
02118, USA
| | - Michael Boehnke
- Center for Statistical Genetics, Department of Biostatistics,
University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Yvonne Boettcher
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Richard S. Cooper
- Department of Public Health Sciences, Stritch School of
Medicine, Loyola University Chicago, Maywood, Illinois
61053, USA
| | - Nita G. Forouhi
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - Christian Gieger
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Harald Grallert
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- German Center for Diabetes Research (DZD),
85764
Neuherberg, Germany
| | - Aroon Hingorani
- Institute of Cardiovascular Science, University College
London, London
WC1E 6BT, UK
| | - Torben Jørgensen
- Department of Clinical Medicine, Faculty of Health and Medical
Sciences, University of Copenhagen, 2200
Copenhagen, Denmark
- Faculty of Medicine, University of Aalborg,
9220
Aalborg, Denmark
- Research Centre for Prevention and Health,
DK2600
Capital Region of Denmark, Denmark
| | - Pekka Jousilahti
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Mika Kivimaki
- Department of Epidemiology and Public Health, UCL,
London
WC1E 6BT, UK
| | - Meena Kumari
- Department of Epidemiology and Public Health, UCL,
London
WC1E 6BT, UK
| | - Markku Laakso
- Faculty of Health Sciences, Institute of Clinical Medicine,
Internal Medicine, University of Eastern Finland, 70210
Kuopio, Finland
- Department of Medicine, University of Eastern Finland,
70210
Kuopio, Finland
- Kuopio University Hospital, 70029
Kuopio, Finland
| | - Claudia Langenberg
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- Department of Epidemiology and Public Health, UCL,
London
WC1E 6BT, UK
| | - Allan Linneberg
- Research Centre for Prevention and Health, Glostrup
Hospital, 2600
Glostrup, Denmark
| | - Amy Luke
- Department of Public Health Sciences, Stritch School of
Medicine, Loyola University Chicago, Maywood, Illinois
61053, USA
| | - Colin A. Mckenzie
- Tropical Metabolism Research Unit, Tropical Medicine Research
Institute, University of the West Indies, Mona
JMAAW15, Jamaica
| | - Aarno Palotie
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
- Massachusetts General Hospital, Center for Human Genetic
Research, Psychiatric and Neurodevelopmental Genetics Unit,
Boston, Massachusetts
02114, USA
| | - Oluf Pedersen
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
| | - Annette Peters
- Institute of Epidemiology II, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Research Unit of Molecular Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
| | - Konstantin Strauch
- Institute of Genetic Epidemiology, Helmholtz Zentrum
München—German Research Center for Environmental
Health, 85764
Neuherberg, Germany
- Institute of Medical Informatics, Biometry and Epidemiology,
Chair of Genetic Epidemiology, Ludwig-Maximilians-Universität,
81377
Munich, Germany
| | - Bamidele O. Tayo
- Department of Public Health Sciences, Stritch School of
Medicine, Loyola University Chicago, Maywood, Illinois
61053, USA
| | - Nicholas J. Wareham
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
| | - David A. Bennett
- Rush Alzheimer's Disease Center, Rush University
Medical Center, Chicago, Illinois
60612, USA
| | - Lars Bertram
- School of Public Health, Faculty of Medicine, Imperial College
London, London
W6 8RP, UK
- Lübeck Interdisciplinary Platform for Genome
Analytics, Institutes of Neurogenetics and Integrative and Experimental
Genomics, University of Lübeck, 23562
Lübeck, Germany
| | - John Blangero
- South Texas Diabetes and Obesity Institute, University of Texas
Rio Grande Valley, Brownsville, Texas
78520
| | - Matthias Blüher
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Claude Bouchard
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - Harry Campbell
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Nam H. Cho
- Ajou University School of Medicine, Department of Preventive
Medicine, Suwon Kyoung-gi
443-721, Korea
| | - Steven R. Cummings
- California Pacific Medical Center Research Institute,
San Francisco, California
94107, USA
| | - Stefan A. Czerwinski
- Lifespan Health Research Center, Wright State University
Boonshoft School of Medicine, Dayton, Ohio
45420, USA
| | - Ilja Demuth
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
- Institute of Medical and Human Genetics,
Charité—Universitätsmedizin Berlin,
13353
Berlin, Germany
| | - Rahel Eckardt
- The Berlin Aging Study II; Research Group on Geriatrics;
Charité—Universitätsmedizin Berlin,
13347
Berlin, Germany
| | - Johan G. Eriksson
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
- Department of General Practice and Primary Health Care,
University of Helsinki, FI-00014
Helsinki, Finland
- Folkhälsan Research Centre, FI-00290
Helsinki, Finland
| | - Luigi Ferrucci
- Translational Gerontology Branch, National Institute on
Aging, Baltimore, Maryland
21225, USA
| | - Oscar H. Franco
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Philippe Froguel
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - Ron T. Gansevoort
- University of Groningen, University Medical Center Groningen,
Department of Medicine, 9700 RB
Groningen, The Netherlands
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Faculty of Health Sciences, University of Southern
Denmark, 5000
Odense, Denmark
| | - Tamara B. Harris
- Laboratory of Epidemiology and Population Sciences, National
Institute on Aging, Bethesda, Maryland
20892, USA
| | - Nicholas Hastie
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Markku Heliövaara
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Albert Hofman
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Joanne M. Jordan
- Thurston Arthritis Research Center, University of North
Carolina at Chapel Hill, Chaper Hill, North Carolina
27599-7280, USA
| | - Antti Jula
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Mika Kähönen
- Department of Clinical Physiology, Tampere University
Hospital, FI-33521
Tampere, Finland
- Department of Clinical Physiology, University of Tampere
School of Medicine, FI-33014
Tampere, Finland
| | - Eero Kajantie
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
- Children's Hospital, Helsinki University Hospital and
University of Helsinki, FI-00029
Helsinki, Finland
- Department of Obstetrics and Gynecology, MRC Oulu, Oulu
University Hospital and University of Oulu, FI-90029
Oulu, Finland
| | - Paul B. Knekt
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Seppo Koskinen
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Peter Kovacs
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
| | - Terho Lehtimäki
- Department of Clinical Chemistry, University of Tampere School
of Medicine, FI-33014
Tampere, Finland
- Department of Clinical Chemistry, Fimlab Laboratories and
School of Medicine, University of Tampere, FI-33520
Tampere, Finland
| | - Lars Lind
- Department of Medical Sciences, Uppsala University,
751 85
Uppsala, Sweden
| | - Yongmei Liu
- Center for Human Genetics, Division of Public Health Sciences,
Wake Forest School of Medicine, Winston-Salem, North
Carolina
27157, USA
| | - Eric S. Orwoll
- Bone & Mineral Unit, Oregon Health & Science
University, Portland, Oregon
97239, USA
| | - Clive Osmond
- MRC Lifecourse Epidemiology Unit, University of Southampton,
Southampton General Hospital, Southampton
SO16 6YD, UK
| | - Markus Perola
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
- Estonian Genome Center, Univeristy of Tartu,
Tartu, 51010, Estonia
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Louis Pérusse
- Department of Kinesiology, Laval University,
Québec City, Quebec, Canada
G1V 0A6
- Institute of Nutrition and Functional Foods, Laval
University, Québec City, Quebec,
Canada
G1V 0A6
| | - Olli T. Raitakari
- Department of Clinical Physiology and Nuclear Medicine, Turku
University Hospital, FI-20521
Turku, Finland
- Research Centre of Applied and Preventive Cardiovascular
Medicine, University of Turku, FI-20520
Turku, Finland
| | - Tuomo Rankinen
- Human Genomics Laboratory, Pennington Biomedical Research
Center, Baton Rouge, Los Angeles
70808, USA
| | - D. C. Rao
- Division of Statistical Genomics, Department of Genetics,
Washington University School of Medicine, St Louis,
Missouri
63108, USA
- Division of Biostatistics, Washington University School of
Medicine, St Louis, Missouri
63110, USA
- Department of Psychiatry, Washington University School of
Medicine, St Louis, Missouri
63110, USA
| | - Treva K. Rice
- Division of Biostatistics, Washington University School of
Medicine, St Louis, Missouri
63110, USA
- Department of Psychiatry, Washington University School of
Medicine, St Louis, Missouri
63110, USA
| | - Fernando Rivadeneira
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Igor Rudan
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Veikko Salomaa
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Thorkild I. A. Sørensen
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
- Institute of Preventive Medicine, Bispebjerg and Frederiksberg
Hospital, The Capital Region, 2000
Frederiksberg, Denmark
| | - Michael Stumvoll
- University of Leipzig, IFB Adiposity Diseases,
04103
Leipzig, Germany
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Anke Tönjes
- University of Leipzig, Department of Medicine,
04103
Leipzig, Germany
| | - Bradford Towne
- Lifespan Health Research Center, Wright State University
Boonshoft School of Medicine, Dayton, Ohio
45420, USA
| | - Gregory J. Tranah
- California Pacific Medical Center Research Institute,
San Francisco, California
94107, USA
| | - Angelo Tremblay
- Department of Kinesiology, Laval University,
Québec City, Quebec, Canada
G1V 0A6
| | - André G. Uitterlinden
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
- Department of Epidemiology, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
| | - Pim van der Harst
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
- Durrer Center for Cardiogenetic Research, Interuniversity
Cardiology Institute Netherlands-Netherlands Heart Institute, 3501
DG
Utrecht, The Netherlands
- Department of Genetics, University Medical Center Groningen,
University of Groningen, 9700 RB
Groningen, The Netherlands
| | - Erkki Vartiainen
- National Institute for Health and Welfare,
FI-00271
Helsinki, Finland
| | - Jorma S. Viikari
- Department of Medicine, University of Turku,
FI-20521
Turku, Finland
| | - Veronique Vitart
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
| | - Marie-Claude Vohl
- Institute of Nutrition and Functional Foods, Laval
University, Québec City, Quebec,
Canada
G1V 0A6
- School of Nutrition, Laval University,
Québec City, Quebec, Canada
G1V 0A6
| | - Henry Völzke
- Institute for Community Medicine, University Medicine
Greifswald, 17475
Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Greifswald, 17475
Greifswald, Germany
- DZD (German Centre for Diabetes Research), partner site
Greifswald, 17475
Greifswald, Germany
| | - Mark Walker
- Program in Medical and Population Genetics, Broad Institute of
Harvard and Massachusetts Institute of Technology, Cambridge,
Massachusetts
02142, USA
- Institute of Cellular Medicine, Newcastle University,
Newcastle
NE2 4HH, UK
| | - Henri Wallaschofski
- Institute of Clinical Chemistry and Laboratory Medicine,
University Medicine Greifswald, 17475
Greifswald, Germany
- DZHK (German Centre for Cardiovascular Research), partner site
Greifswald, 17475
Greifswald, Germany
| | - Sarah Wild
- Centre for Population Health Sciences, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh,
Edinburgh
EH8 9AG, UK
| | - James F. Wilson
- MRC Human Genetics Unit, Institute of Genetics and Molecular
Medicine, University of Edinburgh, Edinburgh
EH4 2XU, UK
- Centre for Global Health Research, Usher Institute of
Population Health Sciences and Informatics, University of Edinburgh, Teviot
Place, Edinburgh
EH8 9AG, UK
| | - Loïc Yengo
- CNRS UMR 8199, F-59019
Lille, France
- European Genomic Institute for Diabetes, 59000
Lille, France
- Université de Lille 2, 59000
Lille, France
| | - D. Timothy Bishop
- Leeds Institute of Cancer and Pathology, Cancer Research UK
Leeds Centre, University of Leeds, Leeds
LS9 7TF, UK
| | - Ingrid B. Borecki
- Division of Statistical Genomics, Department of Genetics,
Washington University School of Medicine, St Louis,
Missouri
63108, USA
- Analytical Genetics Group, Regeneron Genetics Center,
Regeneron Pharmaceuticals, Inc., Tarrytown, New York
10591, USA
| | - John C. Chambers
- Department of Epidemiology and Biostatistics, Imperial College
London, London
W2 1PG, UK
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London
W12 0HS, UK
| | - L. Adrienne Cupples
- Department of Biostatistics, Boston University School of Public
Health, Boston, Massachusetts
02118, USA
- National Heart, Lung, and Blood Institute, the Framingham
Heart Study, Framingham, Massachusetts
01702, USA
| | - Abbas Dehghan
- Department of Epidemiology, Erasmus Medical Center,
3000CA
Rotterdam/Zuidholland, The Netherlands
| | - Panos Deloukas
- William Harvey Research Institute, Barts and The London School
of Medicine and Dentistry, Queen Mary University of London,
London
EC1M 6BQ, UK
- Wellcome Trust Sanger Institute, Human Genetics,
Hinxton, Cambridge
CB10 1SA, UK
- Princess Al-Jawhara Al-Brahim Centre of Excellence in Research
of Hereditary Disorders (PACER-HD), King Abdulaziz University,
Jeddah
21589, Saudi Arabia
| | - Ghazaleh Fatemifar
- MRC Integrative Epidemiology Unit, School of Social and
Community Medicine, University of Bristol, Bristol
BS82BN, UKnited
| | - Caroline Fox
- Harvard Medical School, Boston,
Massachusetts
02115, USA
- National Heart, Lung, and Blood Institute, the Framingham
Heart Study, Framingham, Massachusetts
01702, USA
| | - Terrence S. Furey
- Department of Genetics, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
- Department of Biology, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
| | - Lude Franke
- University of Groningen, University Medical Center Groningen,
Department of Cardiology, 9700 RB
Groningen, The Netherlands
- Department of Genetics, University Medical Center Groningen,
University of Groningen, 9700 RB
Groningen, The Netherlands
| | - Jiali Han
- Department of Epidemiology, Richard M. Fairbanks School of
Public Health, Melvin and Bren Simon Cancer Center,
Indianapolis, Indiana
46202, USA
| | - David J. Hunter
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Department of Epidemiology, Harvard School of Public
Health, Boston, Massachusetts
02115, USA
- Channing Division of Network Medicine, Department of Medicine,
Brigham and Women's Hospital and Harvard Medical School,
Boston, Massachusetts
02115, USA
- Department of Nutrition, Harvard School of Public
Health, Boston, Massachusetts
02115, USA
| | - Juha Karjalainen
- Department of Genetics, University Medical Center Groningen,
University of Groningen, 9700 RB
Groningen, The Netherlands
| | - Fredrik Karpe
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre,
Oxford
OX3 7LJ, UK
| | - Robert C. Kaplan
- Department of Epidemiology and Popualtion Health, Albert
Einstein College of Medicine, Bronx, New York
10461, USA
| | - Jaspal S. Kooner
- Ealing Hospital NHS Trust, Middlesex
UB1 3HW, UK
- Imperial College Healthcare NHS Trust, London
W12 0HS, UK
- National Heart and Lung Institute, Imperial College
London, London
W12 0NN, UK
| | - Mark I. McCarthy
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
- Oxford NIHR Biomedical Research Centre,
Oxford
OX3 7LJ, UK
| | - Joanne M. Murabito
- Boston University School of Medicine, Department of Medicine,
Section of General Internal Medicine, Boston,
Massachusetts
02118, USA
- NHLBI's and Boston University's Framingham
Heart Study, Framingham, Massachusetts
01702, USA
| | - Andrew P. Morris
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Department of Biostatistics, University of Liverpool,
Liverpool
L69 3GA, UK
| | - Julia A. N. Bishop
- Leeds Institute of Cancer and Pathology, Cancer Research UK
Leeds Centre, University of Leeds, Leeds
LS9 7TF, UK
| | - Kari E. North
- Carolina Center for Genome Sciences and Department of
Epidemiology, University of North Carolina at Chapel Hill, Chapel
Hill, North Carolina
27599-7400, USA
| | - Claes Ohlsson
- Centre for Bone and Arthritis Research, Department of Internal
Medicine and Clinical Nutrition, Institute of Medicine, Sahlgrenska Academy,
University of Gothenburg, 413 45
Gothenburg, Sweden
| | - Ken K. Ong
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- MRC Unit for Lifelong Health and Ageing at UCL,
London
WC1B 5JU, UK
- Department of Paediatrics, University of Cambridge,
Cambridge
CB2 0QQ, UK
| | - Inga Prokopenko
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Department of Genomics of Common Disease, School of Public
Health, Imperial College London, London
W12 0NN, UK
- Oxford Centre for Diabetes, Endocrinology and Metabolism,
University of Oxford, Churchill Hospital, Oxford
OX3 7LJ, UK
| | - J. Brent Richards
- Department Epidemiology, Biostatistics and Human Genetics, Lady
Davis Institute, Jewish General Hospital, McGill University,
Montréal, Quebec, Canada
H3T1E2
- Department of Medicine, Lady Davis Institute, Jewish General
Hospital, McGill University, Montréal,
Quebec, Canada
H3T1E2
- Department of Twin Research, King's College
London, London
SE1 1E7, UK
- Division of Endocrinology, Lady Davis Institute, Jewish
General Hospital, McGill University, Montréal,
Quebec, Canada
H3T1E2
| | - Eric E. Schadt
- Icahn Institute for Genomics and Multiscale Biology, Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
- Department of Genetics and Genomic Sciences, Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
| | - Tim D. Spector
- Department of Twin Research and Genetic Epidemiology,
King's College London, London
SE1 7EH, UK
| | - Elisabeth Widén
- Institute for Molecular Medicine Finland, University of
Helsinki, FI-00290
Helsinki, Finland
| | - Cristen J. Willer
- Department of Computational Medicine and Bioinformatics,
University of Michigan, Ann Arbor, Michigan
48109, USA
- Department of Human Genetics, University of Michigan,
Ann Arbor, Michigan
48109, USA
- Department of Internal Medicine, Division of Cardiovascular
Medicine, University of Michigan, Ann Arbor, Michigan
48109, USA
| | - Jian Yang
- Queensland Brain Institute, The University of Queensland,
Brisbane
4072, Australia
| | - Erik Ingelsson
- Science for Life Laboratory, Uppsala University, 750
85
Uppsala, Sweden
- Department of Medical Sciences, Molecular Epidemiology, Uppsala
University, 751 85
Uppsala, Sweden
- Department of Medicine, Division of Cardiovascular Medicine,
Stanford University School of Medicine, Stanford,
California
94305, USA
| | - Karen L. Mohlke
- Department of Genetics, University of North Carolina,
Chapel Hill, North Carolina
27599, USA
| | - Joel N. Hirschhorn
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- Divisions of Endocrinology and Genetics and Center for Basic
and Translational Obesity Research, Boston Children's Hospital,
Boston, Massachusetts
02115, USA
- Department of Genetics, Harvard Medical School,
Boston, Massachusetts
02115, USA
| | - John Andrew Pospisilik
- Department of Epigenetics, Max Planck Institute of
Immunobiology and Epigenetics, D-76108
Freiburg, Germany
| | - M. Carola Zillikens
- Department of Internal Medicine, Erasmus Medical Center,
3015GE
Rotterdam, The Netherlands
- Netherlands Genomics Initiative (NGI)-sponsored Netherlands
Consortium for Healthy Aging (NCHA), Rotterdam
The Netherlands
| | - Cecilia Lindgren
- Wellcome Trust Centre for Human Genetics, University of
Oxford, Oxford
OX3 7BN, UK
- Broad Institute of the Massachusetts Institute of Technology
and Harvard University, Cambridge
2142, USA
- The Big Data Institute, University of Oxford,
Oxford
OX3 7LJ, UK
| | - Tuomas Oskari Kilpeläinen
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- Novo Nordisk Foundation Center for Basic Metabolic Research,
Section of Metabolic Genetics, Faculty of Health and Medical Sciences,
University of Copenhagen, 2100
Copenhagen, Denmark
| | - Ruth J. F. Loos
- The Charles Bronfman Institute for Personalized Medicine, The
Icahn School of Medicine at Mount Sinai, New York, New
York
10029, USA
- The Department of Preventive Medicine, The Icahn School of
Medicine at Mount Sinai, New York, New York
10029, USA
- MRC Epidemiology Unit, University of Cambridge School of
Clinical Medicine, Institute of Metabolic Science, University of Cambridge,
Cambridge Biomedical Campus, Cambridge
CB2 0QQ, UK
- The Genetics of Obesity and Related Metabolic Traits Program,
The Icahn School of Medicine at Mount Sinai, New York, New
York, 10029, USA
- The Mindich Child Health and Development Institute, The Icahn
School of Medicine at Mount Sinai, New York, New York
10029, USA
| |
Collapse
|
50
|
Feng DD, Cai W, Chen X. The associations between Parkinson's disease and cancer: the plot thickens. Transl Neurodegener 2015; 4:20. [PMID: 26504519 PMCID: PMC4620601 DOI: 10.1186/s40035-015-0043-z] [Citation(s) in RCA: 78] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2015] [Accepted: 10/07/2015] [Indexed: 01/10/2023] Open
Abstract
Epidemiological studies support a general inverse association between the risk of cancer development and Parkinson’s disease (PD). In recent years however, increasing amount of eclectic evidence points to a positive association between PD and cancers through different temporal analyses and ethnic groups. This positive association has been supported by several common genetic mutations in SNCA, PARK2, PARK8, ATM, p53, PTEN, and MC1R resulting in cellular changes such as mitochondrial dysfunction, aberrant protein aggregation, and cell cycle dysregulation. Here, we review the epidemiological and biological advances of the past decade in the association between PD and cancers to offer insight on the recent and sometimes contradictory findings.
Collapse
Affiliation(s)
- Danielle D Feng
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA.,Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA USA
| | - Waijiao Cai
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA.,Key Laboratory of Cellular and Molecular Biology, Huashan Hospital, Fudan University, Shanghai, 200040 China
| | - Xiqun Chen
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, MA USA
| |
Collapse
|