1
|
Gokuladhas S, Fadason T, Farrow S, Cooper A, O'Sullivan JM. Discovering genetic mechanisms underlying the co-occurrence of Parkinson's disease and non-motor traits. NPJ Parkinsons Dis 2024; 10:27. [PMID: 38263313 PMCID: PMC10805842 DOI: 10.1038/s41531-024-00638-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/09/2024] [Indexed: 01/25/2024] Open
Abstract
Understanding the biological mechanisms that underlie the non-motor symptoms of Parkinson's disease (PD) requires comprehensive frameworks that unravel the complex interplay of genetic risk factors. Here, we used a disease-agnostic brain cortex gene regulatory network integrated with Mendelian Randomization analyses that identified 19 genes whose changes in expression were causally linked to PD. We further used the network to identify genes that are regulated by PD-associated genome-wide association study (GWAS) SNPs. Extended protein interaction networks derived from PD-risk genes and PD-associated SNPs identified convergent impacts on biological pathways and phenotypes, connecting PD with established co-occurring traits, including non-motor symptoms. These findings hold promise for therapeutic development. In conclusion, while distinct sets of genes likely influence PD risk and outcomes, the existence of genes in common and intersecting pathways associated with other traits suggests that they may contribute to both increased PD risk and symptom heterogeneity observed in people with Parkinson's.
Collapse
Affiliation(s)
- Sreemol Gokuladhas
- The Liggins Institute, University of Auckland, Auckland, 1023, New Zealand
| | - Tayaza Fadason
- The Liggins Institute, University of Auckland, Auckland, 1023, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, 1010, New Zealand
| | - Sophie Farrow
- The Liggins Institute, University of Auckland, Auckland, 1023, New Zealand
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, 1010, New Zealand
| | - Antony Cooper
- St Vincent's Clinical School, UNSW Sydney, Sydney, NSW, Australia
- Australian Parkinson's Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia
| | - Justin M O'Sullivan
- The Liggins Institute, University of Auckland, Auckland, 1023, New Zealand.
- Maurice Wilkins Centre for Molecular Biodiscovery, Auckland, 1010, New Zealand.
- Australian Parkinson's Mission, Garvan Institute of Medical Research, Sydney, New South Wales, Australia.
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton, UK.
- Singapore Institute for Clinical Sciences, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
| |
Collapse
|
2
|
Safi S, Ahmadzade M, Karimi S, Akbari ME, Rouientan H, Abolhosseini M, Rezaei Kanavi M, Khorrami Z. A registration trend in eyelid skin cancers and associated risk factors in Iran, 2005-2016. BMC Cancer 2023; 23:924. [PMID: 37777736 PMCID: PMC10543867 DOI: 10.1186/s12885-023-11414-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2023] [Accepted: 09/17/2023] [Indexed: 10/02/2023] Open
Abstract
BACKGROUND Eyelid skin cancers are the most prevalent ophthalmic malignancies. This study aimed to evaluate the association of the Human Development Index (HDI) and lifestyle risk factors with eyelid skin cancers in Iran. METHODS This ecological study analyzed the data collected from the Iranian National Population-based Cancer Registry (2005-2016). The data on provincial-level eyelid skin cancer risk factors were obtained from national sources. The association between provincial HDI and lifestyle risk factors with the prevalence of eyelid skin cancers was assessed. RESULTS The mean 12-year age-standardized incidence rate (ASIR) of eyelid skin cancers was 16.22 per 100,000 (9,104 cases). The overall ASIR showed an upward trend with an estimated annual average increase of 0.006 per year. There were positive correlations between the prevalence of overall eyelid skin cancers and provincial HDI, smoking, and obesity (r = 0.32, 0.42, and 0.37, respectively). In multivariate analysis, obesity/overweight remained a positive predictor for high prevalence of total eyelid skin cancers (OR = 1.97, 95%CI = 1.08-3.58, P = 0.026), carcinoma (2.10, 1.15-3.83, P = 0.015), and basal cell carcinoma (1.48, 0.99-2.20, P = 0.054). CONCLUSIONS An increasing trend in ASIR of eyelid skin cancers was observed in more than a decade in Iran which was positively associated with provincial HDI and prevalence of obesity. The findings of the study highlight the importance of promotional programs for preventing obesity/overweight and appropriate allocation of screening facilities based on the HDI level.
Collapse
Affiliation(s)
- Sare Safi
- Ophthalmic Epidemiology Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
- Department of Optometry, School of Rehabilitation, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohadese Ahmadzade
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saeed Karimi
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Hamidreza Rouientan
- Ophthalmic Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mohammad Abolhosseini
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Mozhgan Rezaei Kanavi
- Ocular Tissue Engineering Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Zahra Khorrami
- Ophthalmic Epidemiology Research Center, Research Institute for Ophthalmology and Vision Science, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Zhang L, Pozsgai É, Song Y, Macharia J, Alfatafta H, Zheng J, Li Z, Liu H, Kiss I. The relationship between single nucleotide polymorphisms and skin cancer susceptibility: A systematic review and network meta-analysis. Front Oncol 2023; 13:1094309. [PMID: 36874118 PMCID: PMC9975575 DOI: 10.3389/fonc.2023.1094309] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 01/23/2023] [Indexed: 02/17/2023] Open
Abstract
Background Single nucleotide polymorphisms (SNPs) interfere with the function of certain genes and thus may influence the probability of skin cancer. The correlation between SNPs and skin cancer (SC) lacks statistical power, however. Therefore, the purpose of this study was to identify the gene polymorphisms involved in skin cancer susceptibility using network meta-analysis and to determine the relationship between SNPs and SC risk. Methods PubMed, Embase, and Web of Science were searched for articles including "SNP" and different types of SC as keywords between January 2005 and May 2022. The Newcastle-Ottawa Scale was used to assess bias judgments. The odds ratio (ORs) and their 95% confidence intervals (CIs) were determined to estimate heterogeneity within and between studies. Meta-analysis and network meta-analysis were carried out to identify the SNPs associated with SC. The P-score of each SNP was compared to obtain the rank of probability. Subgroup analyses were performed by cancer type. Results A total of 275 SNPs from 59 studies were included in the study. Two subgroup SNP networks using the allele model and dominant model were analyzed. The alternative alleles of rs2228570 (FokI) and rs13181 (ERCC2) were the first-ranking SNPs in both subgroups one and two of the allele model, respectively. The homozygous dominant genotype and heterozygous genotype of rs475007 in subgroup one and the homozygous recessive genotype of rs238406 in subgroup two were most likely to be associated with skin cancer based on the dominant model. Conclusions According to the allele model, SNPs FokI rs2228570 and ERCC2 rs13181 and, according to the dominant model, SNPs MMP1 rs475007 and ERCC2 rs238406 are closely linked to SC risk.
Collapse
Affiliation(s)
- Lu Zhang
- Department of Health Science, Doctoral School of Health Science, University of Pécs, Pécs, Hungary
| | - Éva Pozsgai
- Department of Public Health Medicine, Doctoral School of Clinical Medicine, University of Pécs Medical School, Pécs, Hungary
| | - Yongan Song
- Department of Public Health Medicine, Doctoral School of Clinical Medicine, University of Pécs Medical School, Pécs, Hungary
| | - John Macharia
- Department of Health Science, Doctoral School of Health Science, University of Pécs, Pécs, Hungary
| | - Huda Alfatafta
- Department of Health Science, Doctoral School of Health Science, University of Pécs, Pécs, Hungary
| | - Jia Zheng
- Department of Clinical Epidemiology, the Fourth Affiliated Hospital of China Medical University, Shenyang, China
| | - Zhaoyi Li
- Faculty of Engineering and Information Technology, University of Pécs, Pécs, Hungary
| | - Hongbo Liu
- Department of Health Statistics, School of Public Health, China Medical University, Shenyang, China
| | - István Kiss
- Department of Public Health Medicine, Doctoral School of Clinical Medicine, University of Pécs Medical School, Pécs, Hungary
| |
Collapse
|
4
|
Yardman-Frank JM, Fisher DE. Skin pigmentation and its control: From ultraviolet radiation to stem cells. Exp Dermatol 2020; 30:560-571. [PMID: 33320376 DOI: 10.1111/exd.14260] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In the light of substantial discoveries in epithelial and hair pigmentation pathophysiology, this review summarizes the current understanding of skin pigmentation mechanisms. Melanocytes are pigment-producing cells, and their key regulating transcription factor is the melanocyte-specific microphthalmia-associated transcription factor (m-MITF). Ultraviolet (UV) radiation is a unique modulator of skin pigmentation influencing tanning pathways. The delayed tanning pathway occurs as UVB produces keratinocyte DNA damage, causing p53-mediated expression of the pro-opiomelanocortin (POMC) gene that is processed to release α-melanocyte-stimulating hormone (α-MSH). α-MSH stimulates the melanocortin 1 receptor (MC1R) on melanocytes, leading to m-MITF expression and melanogenesis. POMC cleavage also releases β-endorphin, which creates a neuroendocrine pathway that promotes UV-seeking behaviours. Mutations along the tanning pathway can affect pigmentation and increase the risk of skin malignancies. MC1R variants have received considerable attention, yet the allele is highly polymorphic with varied phenotypes. Vitiligo presents with depigmented skin lesions due to autoimmune destruction of melanocytes. UVB phototherapy stimulates melanocyte stem cells in the hair bulge to undergo differentiation and upwards migration resulting in perifollicular repigmentation of vitiliginous lesions, which is under sophisticated signalling control. Melanocyte stem cells, normally quiescent, undergo cyclic activation/differentiation and downward migration with the hair cycle, providing pigment to hair follicles. Physiological hair greying results from progressive loss of melanocyte stem cells and can be accelerated by acute stress-induced, sympathetic driven hyperproliferation of the melanocyte stem cells. Ultimately, by reviewing the pathways governing epithelial and follicular pigmentation, numerous areas of future research and potential points of intervention are highlighted.
Collapse
Affiliation(s)
| | - David E Fisher
- Department of Dermatology, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
5
|
Harman RM, Das SP, Bartlett AP, Rauner G, Donahue LR, Van de Walle GR. Beyond tradition and convention: benefits of non-traditional model organisms in cancer research. Cancer Metastasis Rev 2020; 40:47-69. [PMID: 33111160 DOI: 10.1007/s10555-020-09930-6] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 09/01/2020] [Indexed: 02/07/2023]
Abstract
Traditional laboratory model organisms are indispensable for cancer research and have provided insight into numerous mechanisms that contribute to cancer development and progression in humans. However, these models do have some limitations, most notably related to successful drug translation, because traditional model organisms are often short-lived, small-bodied, genetically homogeneous, often immunocompromised, are not exposed to natural environments shared with humans, and usually do not develop cancer spontaneously. We propose that assimilating information from a variety of long-lived, large, genetically diverse, and immunocompetent species that live in natural environments and do develop cancer spontaneously (or do not develop cancer at all) will lead to a more comprehensive understanding of human cancers. These non-traditional model organisms can also serve as sentinels for environmental risk factors that contribute to human cancers. Ultimately, expanding the range of animal models that can be used to study cancer will lead to improved insights into cancer development, progression and metastasis, tumor microenvironment, as well as improved therapies and diagnostics, and will consequently reduce the negative impacts of the wide variety of cancers afflicting humans overall.
Collapse
Affiliation(s)
- Rebecca M Harman
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Sanjna P Das
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Arianna P Bartlett
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gat Rauner
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Leanne R Donahue
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA
| | - Gerlinde R Van de Walle
- Baker Institute for Animal Health, College of Veterinary Medicine, Cornell University, Ithaca, NY, 14853, USA.
| |
Collapse
|
6
|
Abstract
Human skin and hair color are visible traits that can vary dramatically within and across ethnic populations. The genetic makeup of these traits-including polymorphisms in the enzymes and signaling proteins involved in melanogenesis, and the vital role of ion transport mechanisms operating during the maturation and distribution of the melanosome-has provided new insights into the regulation of pigmentation. A large number of novel loci involved in the process have been recently discovered through four large-scale genome-wide association studies in Europeans, two large genetic studies of skin color in Africans, one study in Latin Americans, and functional testing in animal models. The responsible polymorphisms within these pigmentation genes appear at different population frequencies, can be used as ancestry-informative markers, and provide insight into the evolutionary selective forces that have acted to create this human diversity.
Collapse
Affiliation(s)
- William J Pavan
- Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892, USA;
| | - Richard A Sturm
- Dermatology Research Centre, The University of Queensland Diamantina Institute, The University of Queensland, Brisbane, Queensland 4102, Australia;
| |
Collapse
|
7
|
Fesenko DO, Abramov IS, Shershov VE, Kuznetsova VE, Surzhikov SA, Grechishnikova IV, Barsky VE, Chudinov AV, Nasedkina TV. Multiplex Assay to Evaluate the Genetic Risk of Developing Human Melanoma. Mol Biol 2018. [DOI: 10.1134/s0026893318060079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
8
|
Nair-Shalliker V, Egger S, Chrzanowska A, Mason R, Waite L, Le Couteur D, Seibel MJ, Handelsman DJ, Cumming R, Smith DP, Armstrong BK. Associations between sun sensitive pigmentary genes and serum prostate specific antigen levels. PLoS One 2018. [PMID: 29518100 PMCID: PMC5843239 DOI: 10.1371/journal.pone.0193893] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Background Melanoma and prostate cancer may share risk factors. This study examined the association between serum PSA levels, which is a risk factor for prostate cancer, and variants in some melanoma-associated pigmentary genes. Methods We studied participants, all aged 70+ years, in the Concord Health and Ageing in Men Project who had no history of prostatitis or received treatment for prostate disease (n = 1033). We genotyped variants in MC1R (rs1805007, rs1805008), ASIP (rs4911414, rs1015362), SLC45A2 (rs28777, rs16891982), IRF4 (rs12203592), TYRP1 (rs1408799), TYR (rs1126809, rs1042602), SLC24A2 (rs12896399), and OCA2 (rs7495174). Generalised linear dominant models with Poisson distribution, log link functions and robust variance estimators estimated adjusted percentage differences (%PSA) in mean serum PSA levels (ng/mL) between variant and wildtype (0%PSA = reference) genotypes, adjusting for age, body mass index, serum 25OHD levels and birth regions (Australia or New Zealand (ANZ), Europe or elsewhere). Results Serum PSA levels were strongly associated with advancing age and birth regions: mean PSA levels were lower in Europe-born (-29.7%) and elsewhere-born (-11.7%) men than ANZ-born men (reference). Lower %PSA was observed in men with variants in SLC45A2: rs28777 (-19.6;95%CI: -33.5, -2.7), rs16891982 (-17.3;95%CI:-30.4,-1.7) than in wildtype men (reference). There were significant interactions between birth regions and PSA levels in men with variants in MC1R (rs1805007; p-interaction = 0.0001) and ASIP (rs4911414; p-interaction = 0.007). For these genes %PSA was greater in ANZ-born men and lower in Europe- and elsewhere-born men with the variant than it was in wildtype men. In a post hoc analysis, serum testosterone levels were increased in men with MC1R rs1805007 and serum dihydrotestosterone in men with ASIP rs1015362. Conclusion Men with SNPs in SLC45A2, who have less sun sensitive skin, have lower PSA levels. Men with SNPs in MC1R and ASIP, who have more sun sensitive skin, and were born in ANZ, have higher PSA levels. Androgens may modify these apparent associations of pigmentary genes and sun exposure with PSA levels. Impact PSA levels and possibly prostate cancer risk may vary with sun sensitivity and sun exposure, the effects of which might be modified by androgen levels.
Collapse
Affiliation(s)
- Visalini Nair-Shalliker
- Cancer Research Division, Cancer Council New South Wales, Sydney, Australia
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Department of Clinical Medicine, Macquarie University, Sydney, Australia
- * E-mail:
| | - Sam Egger
- Cancer Research Division, Cancer Council New South Wales, Sydney, Australia
| | - Agata Chrzanowska
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
| | - Rebecca Mason
- Sydney Medical School, The University of Sydney, Sydney, Australia
| | - Louise Waite
- Centre for Education and Research on Ageing, Concord Hospital and The University of Sydney, Sydney, New South Wales, Australia
| | - David Le Couteur
- Centre for Education and Research on Ageing, Concord Hospital and The University of Sydney, Sydney, New South Wales, Australia
| | - Markus J. Seibel
- Centre for Education and Research on Ageing, Concord Hospital and The University of Sydney, Sydney, New South Wales, Australia
| | - David J. Handelsman
- Centre for Education and Research on Ageing, Concord Hospital and The University of Sydney, Sydney, New South Wales, Australia
| | - Robert Cumming
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Centre for Education and Research on Ageing, Concord Hospital and The University of Sydney, Sydney, New South Wales, Australia
- ANZAC Research Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - David P. Smith
- Cancer Research Division, Cancer Council New South Wales, Sydney, Australia
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- Menzies Health Institute Queensland, Griffith University, Southport, Queensland, Australia
| | - Bruce K. Armstrong
- Sydney School of Public Health, The University of Sydney, Sydney, Australia
- School of Population Health, University of Western Australia, Perth, Western Australia, Australia
| |
Collapse
|
9
|
Kansal RG, McCravy MS, Basham JH, Earl JA, McMurray SL, Starner CJ, Whitt MA, Albritton LM. Inhibition of melanocortin 1 receptor slows melanoma growth, reduces tumor heterogeneity and increases survival. Oncotarget 2018; 7:26331-45. [PMID: 27028866 PMCID: PMC5041983 DOI: 10.18632/oncotarget.8372] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2016] [Accepted: 03/14/2016] [Indexed: 11/25/2022] Open
Abstract
Melanoma risk is increased in patients with mutations of melanocortin 1 receptor (MC1R) yet the basis for the increased risk remains unknown. Here we report in vivo evidence supporting a critical role for MC1R in regulating melanoma tumor growth and determining overall survival time. Inhibition of MC1R by its physiologically relevant competitive inhibitor, agouti signaling protein (ASIP), reduced melanin synthesis and morphological heterogeneity in murine B16-F10 melanoma cells. In the lungs of syngeneic C57BL/6 mice, mCherry-marked, ASIP-secreting lung tumors inhibited MC1R on neighboring tumors lacking ASIP in a dose dependent manner as evidenced by a proportional loss of pigment in tumors from mice injected with 1:1, 3:1 and 4:1 mixtures of parental B16-F10 to ASIP-expressing tumor cells. ASIP-expressing B16-F10 cells formed poorly pigmented tumors in vivo that correlated with a 20% longer median survival than those bearing parental B16-F10 tumors (p=0.0005). Mice injected with 1:1 mixtures also showed survival benefit (p=0.0054), whereas injection of a 4:1 mixture showed no significant difference in survival. The longer survival time of mice bearing ASIP-expressing tumors correlated with a significantly slower growth rate than parental B16-F10 tumors as judged by quantification of numbers of tumors and total tumor load (p=0.0325), as well as a more homogeneous size and morphology of ASIP-expressing lung tumors. We conclude that MC1R plays an important role in regulating melanoma growth and morphology. Persistent inhibition of MC1R provided a significant survival advantage resulting in part from slower tumor growth, establishing MC1R as a compelling new molecular target for metastatic melanoma.
Collapse
Affiliation(s)
- Rita G Kansal
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Matthew S McCravy
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Jacob H Basham
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Joshua A Earl
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Stacy L McMurray
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Chelsey J Starner
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Michael A Whitt
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| | - Lorraine M Albritton
- Department of Microbiology, Immunology and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, TN 38163, USA
| |
Collapse
|
10
|
Clement E, Lazar I, Muller C, Nieto L. Obesity and melanoma: could fat be fueling malignancy? Pigment Cell Melanoma Res 2017; 30:294-306. [PMID: 28222242 DOI: 10.1111/pcmr.12584] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2016] [Accepted: 02/13/2017] [Indexed: 01/01/2023]
Abstract
Over the last decade, it has become increasingly clear that adipose tissue, and particularly adipocytes, contributes to tumor progression. Obesity, an ever-increasing worldwide phenomenon, exacerbates this effect. The influence of obesity on melanoma remains poorly studied, although recent data do underline an association between the two diseases in both humans and murine models. Herein, we review the impact of obesity on melanoma incidence and progression and discuss the underlying mechanisms known to be involved. Adipose tissue favors the proliferation and aggressiveness of melanoma cells through a direct dialog, mediated by soluble factors and by exosomes, and through remodeling of the tumor microenvironment. This knowledge could, in the future, help to design new personalized therapeutic options for obese melanoma patients.
Collapse
Affiliation(s)
- Emily Clement
- Université de Toulouse, CNRS, UPS, Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse Cedex, France
| | - Ikrame Lazar
- Université de Toulouse, CNRS, UPS, Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse Cedex, France
| | - Catherine Muller
- Université de Toulouse, CNRS, UPS, Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse Cedex, France
| | - Laurence Nieto
- Université de Toulouse, CNRS, UPS, Institut de Pharmacologie et de Biologie Structurale (IPBS), Toulouse Cedex, France
| |
Collapse
|
11
|
Mobuchon L, Battistella A, Bardel C, Scelo G, Renoud A, Houy A, Cassoux N, Milder M, Cancel-Tassin G, Cussenot O, Delattre O, Besse C, Boland A, Deleuze JF, Cox DG, Stern MH. A GWAS in uveal melanoma identifies risk polymorphisms in the CLPTM1L locus. NPJ Genom Med 2017; 2:5. [PMID: 28781888 PMCID: PMC5542017 DOI: 10.1038/s41525-017-0008-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Revised: 01/31/2017] [Accepted: 02/01/2017] [Indexed: 02/03/2023] Open
Abstract
Uveal melanoma, a rare malignant tumor of the eye, is predominantly observed in populations of European ancestry. A genome-wide association study of 259 uveal melanoma patients compared to 401 controls all of European ancestry revealed a candidate locus at chromosome 5p15.33 (region rs421284: OR = 1.7, CI 1.43-2.05). This locus was replicated in an independent set of 276 cases and 184 controls. In addition, risk variants from this region were positively associated with higher expression of CLPTM1L. In conclusion, the CLPTM1L region contains risk alleles for uveal melanoma susceptibility, suggesting that CLPTM1L could play a role in uveal melanoma oncogenesis.
Collapse
Affiliation(s)
- Lenha Mobuchon
- Inserm U830 and Ensemble Hospitalier, PSL Research University, Institut Curie, Paris, France
| | - Aude Battistella
- Inserm U830 and Ensemble Hospitalier, PSL Research University, Institut Curie, Paris, France
| | - Claire Bardel
- UMR5558, Laboratoire de Biométrie et Biologie Evolutive, Equipe Biostatistique-Santé, Université Claude Bernard-Lyon 1, Lyon, France
- Service de Biostatistique-bioinformatique, Hospices Civils de Lyon, Lyon, France
| | - Ghislaine Scelo
- International Agency for Research on Cancer (IARC), Lyon, France
| | - Alexia Renoud
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Alexandre Houy
- Inserm U830 and Ensemble Hospitalier, PSL Research University, Institut Curie, Paris, France
| | - Nathalie Cassoux
- Inserm U830 and Ensemble Hospitalier, PSL Research University, Institut Curie, Paris, France
| | - Maud Milder
- Inserm U830 and Ensemble Hospitalier, PSL Research University, Institut Curie, Paris, France
| | | | - Olivier Cussenot
- UPMC University Paris 06 GRC n°5, CeRePP, Hôpital Tenon, Paris, France
| | - Olivier Delattre
- Inserm U830 and Ensemble Hospitalier, PSL Research University, Institut Curie, Paris, France
| | - Céline Besse
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | - Anne Boland
- Centre National de Génotypage, Institut de Génomique, CEA, Evry, France
| | | | - David G. Cox
- INSERM U1052, CNRS UMR5286, Université Lyon 1, Centre de Recherche en Cancérologie de Lyon, Lyon, France
| | - Marc-Henri Stern
- Inserm U830 and Ensemble Hospitalier, PSL Research University, Institut Curie, Paris, France
| |
Collapse
|
12
|
Asgari MM, Wang W, Ioannidis NM, Itnyre J, Hoffmann T, Jorgenson E, Whittemore AS. Identification of Susceptibility Loci for Cutaneous Squamous Cell Carcinoma. J Invest Dermatol 2016; 136:930-937. [PMID: 26829030 DOI: 10.1016/j.jid.2016.01.013] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2015] [Revised: 01/04/2016] [Accepted: 01/06/2016] [Indexed: 12/20/2022]
Abstract
We report a genome-wide association study of cutaneous squamous cell carcinoma conducted among non-Hispanic white members of the Kaiser Permanente Northern California health care system. The study includes a genome-wide screen of 61,457 members (6,891 cases and 54,566 controls) genotyped on the Affymetrix Axiom European array and a replication phase involving an independent set of 6,410 additional members (810 cases and 5,600 controls). Combined analysis of screening and replication phases identified 10 loci containing single-nucleotide polymorphisms (SNPs) with P-values < 5 × 10(-8). Six loci contain genes in the pigmentation pathway; SNPs at these loci appear to modulate squamous cell carcinoma risk independently of the pigmentation phenotypes. Another locus contains HLA class II genes studied in relation to elevated squamous cell carcinoma risk following immunosuppression. SNPs at the remaining three loci include an intronic SNP in FOXP1 at locus 3p13, an intergenic SNP at 3q28 near TP63, and an intergenic SNP at 9p22 near BNC2. These findings provide insights into the genetic factors accounting for inherited squamous cell carcinoma susceptibility.
Collapse
Affiliation(s)
- Maryam M Asgari
- Department of Dermatology, Massachusetts General Hospital, Boston, Massachusetts, USA; Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Wei Wang
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California, USA
| | - Nilah M Ioannidis
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California, USA; Department of Genetics, Stanford University School of Medicine, Stanford, California, USA
| | - Jacqueline Itnyre
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California, USA
| | - Thomas Hoffmann
- Department of Epidemiology and Biostatistics and Institute for Human Genetics, University of California, San Francisco, California, USA
| | - Eric Jorgenson
- Division of Research, Kaiser Permanente Northern California, Oakland, California, USA
| | - Alice S Whittemore
- Department of Health Research and Policy, Stanford University School of Medicine, Stanford, California, USA.
| |
Collapse
|
13
|
Abstract
Melanoma is increasing in incidence and represents an aggressive type of cancer. Efforts have focused on identifying genetic factors in melanoma carcinogenesis to guide prevention, screening, early detection, and targeted therapy. This article reviews the hereditary risk factors associated with melanoma and the known molecular pathways and genetic mutations associated with this disease. This article also explores the controversies associated with genetic testing and the latest advances in identifying genetic targets in melanoma, which offer promise for future application in the multidisciplinary management of melanoma.
Collapse
Affiliation(s)
- Omar M Rashid
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, SRB 4.24012, Tampa, FL 33612, USA; Bienes Comprehensive Cancer Center, Holy Cross Hospital, 4725 N Federal Highway, Fort Lauderdale, FL 33308, USA
| | - Jonathan S Zager
- Department of Cutaneous Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, SRB 4.24012, Tampa, FL 33612, USA.
| |
Collapse
|
14
|
Taylor NJ, Reiner AS, Begg CB, Cust AE, Busam KJ, Anton-Culver H, Dwyer T, From L, Gallagher RP, Gruber SB, Rosso S, White KA, Zanetti R, Orlow I, Thomas NE, Rebbeck TR, Berwick M, Kanetsky PA. Inherited variation at MC1R and ASIP and association with melanoma-specific survival. Int J Cancer 2014; 136:2659-67. [PMID: 25382380 DOI: 10.1002/ijc.29317] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 10/22/2014] [Indexed: 01/04/2023]
Abstract
Melanocortin-1 receptor (MC1R) is a marker of melanoma risk in populations of European ancestry. However, MC1R effects on survival are much less studied. We investigated associations between variation at MC1R and survival in an international, population-based series of single primary melanoma patients enrolled into the Genes, Environment, and Melanoma study. MC1R genotype data was available for 2,200 participants with a first incident primary melanoma diagnosis. We estimated the association of MC1R genotypes with melanoma-specific survival (i.e., death caused by melanoma) and overall survival using COX proportional hazards modeling, adjusting for established prognostic factors for melanoma. We also conducted stratified analyses by Breslow thickness, tumor site, phenotypic index, and age. In addition, we evaluated haplotypes involving polymorphisms near the Agouti signaling protein gene (ASIP) locus for their impacts on survival. Melanoma-specific survival was inversely associated with carriage of MC1R variants in the absence of consensus alleles compared to carriage of at least one consensus allele (hazard ratio (HR) = 0.60; 95% confidence interval (CI): 0.40, 0.90). MC1R results for overall survival were consistent with no association. We did not observe any statistical evidence of heterogeneity of effect estimates in stratified analyses. We observed increased hazard of melanoma-specific death among carriers of the risk haplotype TG near the ASIP locus (HR = 1.37; 95% CI: 0.91, 2.04) when compared to carriers of the most common GG haplotype. Similar results were noted for overall survival. Upon examining the ASIP TG/TG diplotype, we observed considerably increased hazard of melanoma-specific death (HR = 5.11; 95% CI: 1.88, 13.88) compared to carriers of the most common GG/GG diplotype. Our data suggest improved melanoma-specific survival among carriers of two inherited MC1R variants.
Collapse
Affiliation(s)
- Nicholas J Taylor
- Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Liu-Smith F, Poe C, Farmer PJ, Meyskens FL. Amyloids, melanins and oxidative stress in melanomagenesis. Exp Dermatol 2014; 24:171-4. [PMID: 25271672 DOI: 10.1111/exd.12559] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/24/2014] [Indexed: 12/26/2022]
Abstract
Melanoma has traditionally been viewed as an ultraviolet (UV) radiation-induced malignancy. While UV is a common inducing factor, other endogenous stresses such as metal ion accumulation or the melanin pigment itself may provide alternative pathways to melanoma progression. Eumelanosomes within melanoma often exhibit disrupted membranes and fragmented pigment which may be due to alterations in their amyloid-based striated matrix. The melanosomal amyloid can itself be toxic, especially in combination with reactive oxygen species (ROS) and reactive nitrogen species (RNS) generated by endogenous NADPH oxidase (NOX) and nitric oxide synthase (NOS) enzymes, a toxic mix that may initiate melanomagenesis. Further understanding of the loss of the melanosomal organization, the behaviour of the exposed melanin and the induction of ROS/RNS in melanomas may provide critical insights into this deadly disease.
Collapse
Affiliation(s)
- Feng Liu-Smith
- Department of Epidemiology, University of California School of Medicine, Irvine, CA, USA; Department of Medicine, University of California School of Medicine, Irvine, CA, USA; Chao Family Comprehensive Cancer Center, University of California School of Medicine, Irvine, CA, USA
| | | | | | | |
Collapse
|
16
|
García-Borrón JC, Abdel-Malek Z, Jiménez-Cervantes C. MC1R, the cAMP pathway, and the response to solar UV: extending the horizon beyond pigmentation. Pigment Cell Melanoma Res 2014; 27:699-720. [PMID: 24807163 DOI: 10.1111/pcmr.12257] [Citation(s) in RCA: 140] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Accepted: 05/01/2014] [Indexed: 12/20/2022]
Abstract
The melanocortin 1 receptor (MC1R) is a G protein-coupled receptor crucial for the regulation of melanocyte proliferation and function. Upon binding melanocortins, MC1R activates several signaling cascades, notably the cAMP pathway leading to synthesis of photoprotective eumelanin. Polymorphisms in the MC1R gene are a major source of normal variation of human hair color and skin pigmentation, response to ultraviolet radiation (UVR), and skin cancer susceptibility. The identification of a surprisingly high number of MC1R natural variants strongly associated with pigmentary phenotypes and increased skin cancer risk has prompted research on the functional properties of the wild-type receptor and frequent mutant alleles. We summarize current knowledge on MC1R structural and functional properties, as well as on its intracellular trafficking and signaling. We also review the current knowledge about the function of MC1R as a skin cancer, particularly melanoma, susceptibility gene and how it modulates the response of melanocytes to UVR.
Collapse
Affiliation(s)
- Jose C García-Borrón
- Department of Biochemistry and Molecular Biology, School of Medicine, University of Murcia, Murcia, Spain; Instituto Murciano de Investigación Biomédica (IMIB), El Palmar, Murcia, Spain
| | | | | |
Collapse
|
17
|
Shi J, Yang XR, Ballew B, Rotunno M, Calista D, Fargnoli MC, Ghiorzo P, Bressac-de Paillerets B, Nagore E, Avril MF, Caporaso NE, McMaster ML, Cullen M, Wang Z, Zhang X, Bruno W, Pastorino L, Queirolo P, Banuls-Roca J, Garcia-Casado Z, Vaysse A, Mohamdi H, Riazalhosseini Y, Foglio M, Jouenne F, Hua X, Hyland PL, Yin J, Vallabhaneni H, Chai W, Minghetti P, Pellegrini C, Ravichandran S, Eggermont A, Lathrop M, Peris K, Scarra GB, Landi G, Savage SA, Sampson JN, He J, Yeager M, Goldin LR, Demenais F, Chanock SJ, Tucker MA, Goldstein AM, Liu Y, Landi MT. Rare missense variants in POT1 predispose to familial cutaneous malignant melanoma. Nat Genet 2014; 46:482-486. [PMID: 24686846 PMCID: PMC4056593 DOI: 10.1038/ng.2941] [Citation(s) in RCA: 248] [Impact Index Per Article: 22.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2013] [Accepted: 03/07/2014] [Indexed: 12/15/2022]
Abstract
Although CDKN2A is the most frequent high-risk melanoma susceptibility gene, the underlying genetic factors for most melanoma-prone families remain unknown. Using whole-exome sequencing, we identified a rare variant that arose as a founder mutation in the telomere shelterin gene POT1 (chromosome 7, g.124493086C>T; p.Ser270Asn) in five unrelated melanoma-prone families from Romagna, Italy. Carriers of this variant had increased telomere lengths and numbers of fragile telomeres, suggesting that this variant perturbs telomere maintenance. Two additional rare POT1 variants were identified in all cases sequenced in two separate Italian families, one variant per family, yielding a frequency for POT1 variants comparable to that for CDKN2A mutations in this population. These variants were not found in public databases or in 2,038 genotyped Italian controls. We also identified two rare recurrent POT1 variants in US and French familial melanoma cases. Our findings suggest that POT1 is a major susceptibility gene for familial melanoma in several populations.
Collapse
Affiliation(s)
- Jianxin Shi
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2]
| | - Xiaohong R Yang
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2]
| | - Bari Ballew
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Melissa Rotunno
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Donato Calista
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | | | - Paola Ghiorzo
- 1] Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy. [2] Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | | | - Eduardo Nagore
- 1] Department of Dermatology, Instituto Valenciano de Oncología, Valencia, Spain. [2] Department of Dermatology, Universidad Católica de Valencia, Valencia, Spain
| | - Marie Francoise Avril
- Université Paris Descartes, Assistance Publique-Hôpitaux de Paris (AP-HP), Hôpital Cochin, Paris, France
| | - Neil E Caporaso
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Mary L McMaster
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Michael Cullen
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Zhaoming Wang
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Xijun Zhang
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - William Bruno
- 1] Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy. [2] Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Lorenza Pastorino
- 1] Department of Internal Medicine and Medical Specialties, University of Genoa, Genoa, Italy. [2] Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Paola Queirolo
- Genetics of Rare Hereditary Cancers, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) San Martino-IST Istituto Nazionale per la Ricerca sul Cancro, Genoa, Italy
| | - Jose Banuls-Roca
- Department of Dermatology, Hospital General Universitario de Alicante, Alicante, Spain
| | - Zaida Garcia-Casado
- Laboratory of Molecular Biology, Instituto Valenciano de Oncología, Valencia, Spain
| | - Amaury Vaysse
- 1] INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France. [2] Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| | - Hamida Mohamdi
- 1] INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France. [2] Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| | - Yasser Riazalhosseini
- 1] McGill University and Génome Québec Innovation Centre, Montreal, Quebec, Canada. [2] Department of Human Genetics, McGill University, Montreal, Quebec, Canada
| | | | | | - Xing Hua
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Paula L Hyland
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Jinhu Yin
- Laboratory of Molecular Gerontology, National Institute on Aging, US National Institutes of Health, US Department of Health and Human Services, Baltimore, Maryland, USA
| | - Haritha Vallabhaneni
- Laboratory of Molecular Gerontology, National Institute on Aging, US National Institutes of Health, US Department of Health and Human Services, Baltimore, Maryland, USA
| | - Weihang Chai
- Section of Medical Sciences, School of Molecular Biosciences, Washington State University, Spokane, Washington, USA
| | - Paola Minghetti
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Cristina Pellegrini
- Department of Biotechnological and Applied Clinical Sciences, University of L'Aquila, L'Aquila, Italy
| | - Sarangan Ravichandran
- SAIC-Frederick, Inc., Frederick National Laboratory for Cancer Research, Simulation, Analysis and Mathematical Modeling Group, Advanced Biomedical Computing Center, Frederick, Maryland, USA
| | - Alexander Eggermont
- 1] Service de Génétique, Gustave Roussy, Villejuif, France. [2] Université Paris-Sud, Kremlin Bicêtre France, Gustave Roussy, Villejuif, France
| | - Mark Lathrop
- 1] McGill University and Génome Québec Innovation Centre, Montreal, Quebec, Canada. [2] Department of Human Genetics, McGill University, Montreal, Quebec, Canada. [3] Fondation Jean Dausset-Centre d'Etude du Polymorphisme Humain (CEPH), Paris, France
| | - Ketty Peris
- Department of Dermatology, University of L'Aquila, L'Aquila, Italy
| | | | - Giorgio Landi
- Department of Dermatology, Maurizio Bufalini Hospital, Cesena, Italy
| | - Sharon A Savage
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Joshua N Sampson
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Ji He
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Meredith Yeager
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2] Cancer Genomics Research Laboratory, NCI-Frederick, SAIC-Frederick, Inc., Frederick, Maryland, USA
| | - Lynn R Goldin
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Florence Demenais
- 1] INSERM, UMR 946, Genetic Variation and Human Diseases Unit, Paris, France. [2] Université Paris Diderot, Sorbonne Paris Cité, Institut Universitaire d'Hématologie, Paris, France
| | - Stephen J Chanock
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Margaret A Tucker
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Alisa M Goldstein
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA
| | - Yie Liu
- Laboratory of Molecular Gerontology, National Institute on Aging, US National Institutes of Health, US Department of Health and Human Services, Baltimore, Maryland, USA
| | - Maria Teresa Landi
- 1] Division of Cancer Epidemiology and Genetics, National Cancer Institute, US National Institutes of Health, US Department of Health and Human Services, Bethesda, Maryland, USA. [2]
| |
Collapse
|
18
|
Liu-Smith F, Dellinger R, Meyskens FL. Updates of reactive oxygen species in melanoma etiology and progression. Arch Biochem Biophys 2014; 563:51-5. [PMID: 24780245 DOI: 10.1016/j.abb.2014.04.007] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Revised: 04/14/2014] [Accepted: 04/17/2014] [Indexed: 01/01/2023]
Abstract
Reactive oxygen species (ROS) play crucial roles in all aspects of melanoma development, however, the source of ROS is not well defined. In this review we summarize recent advancement in this rapidly developing field. The cellular ROS pool in melanocytes can be derived from mitochondria, melanosomes, NADPH oxidase (NOX) family enzymes, and uncoupling of nitric oxide synthase (NOS). Current evidence suggests that Nox1, Nox4 and Nox5 are expressed in melanocytic lineage. While there is no difference in Nox1 expression levels in primary and metastatic melanoma tissues, Nox4 expression is significantly higher in a subset of metastatic melanoma tumors as compared to the primary tumors; suggesting distinct and specific signals and effects for NOX family enzymes in melanoma. Targeting these NOX enzymes using specific NOX inhibitors may be effective for a subset of certain tumors. ROS also play important roles in BRAF inhibitor induced drug resistance; hence identification and blockade of the source of this ROS may be an effective way to enhance efficacy and overcome resistance. Furthermore, ROS from different sources may interact with each other and interact with reactive nitrogen species (RNS) and drive the melanomagenesis process at all stages of disease. Further understanding ROS and RNS in melanoma etiology and progression is necessary for developing new prevention and therapeutic approaches.
Collapse
Affiliation(s)
- Feng Liu-Smith
- Department of Epidemiology, University of California School of Medicine, Irvine, CA 92697, United States; Department of Medicine, University of California School of Medicine, Irvine, CA 92697, United States; Chao Family Comprehensive Cancer Center, University of California School of Medicine, Irvine, CA 92697, United States.
| | - Ryan Dellinger
- Department of Medicine, University of California School of Medicine, Irvine, CA 92697, United States; Chao Family Comprehensive Cancer Center, University of California School of Medicine, Irvine, CA 92697, United States
| | - Frank L Meyskens
- Department of Epidemiology, University of California School of Medicine, Irvine, CA 92697, United States; Department of Medicine, University of California School of Medicine, Irvine, CA 92697, United States; Department of Biological Chemistry, University of California School of Medicine, Irvine, CA 92697, United States; Department of Public Health, University of California School of Medicine, Irvine, CA 92697, United States; Chao Family Comprehensive Cancer Center, University of California School of Medicine, Irvine, CA 92697, United States
| |
Collapse
|
19
|
Hill VK, Gartner JJ, Samuels Y, Goldstein AM. The genetics of melanoma: recent advances. Annu Rev Genomics Hum Genet 2013; 14:257-79. [PMID: 23875803 DOI: 10.1146/annurev-genom-091212-153429] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Cutaneous malignant melanoma results from the interplay of genetic, host, and environmental factors. Genetic factors implicated in melanoma etiology include inherited high-, intermediate-, and low-risk susceptibility genes as well as numerous somatic mutations in melanoma tumors. CDKN2A is the major high-risk melanoma susceptibility gene identified to date. Recent identification of low-risk loci has been accomplished predominantly through genome-wide association studies. Whole-exome and whole-genome studies have identified numerous genes somatically altered in melanoma tumors and highlighted a higher mutation load in melanoma tumors compared with those in other cancers. This higher load is believed to be attributable to the preponderance of cytosine-to-thymine nucleotide substitutions as a result of UV radiation exposure. Technological advances, particularly next-generation sequencing, have increased the opportunities for germline and somatic gene discovery in melanoma and are opening up new avenues for understanding melanoma pathogenesis as well as leading to new opportunities for treatment.
Collapse
Affiliation(s)
- Victoria K Hill
- Cancer Genetics Branch, National Human Genome Research Institute, National Institutes of Health, Bethesda, Maryland 20892
| | | | | | | |
Collapse
|
20
|
Maccioni L, Rachakonda PS, Bermejo JL, Planelles D, Requena C, Hemminki K, Nagore E, Kumar R. Variants at the 9p21 locus and melanoma risk. BMC Cancer 2013; 13:325. [PMID: 23816148 PMCID: PMC3702420 DOI: 10.1186/1471-2407-13-325] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2013] [Accepted: 06/27/2013] [Indexed: 12/16/2022] Open
Abstract
Background The influence of variants at the 9p21 locus on melanoma risk has been reported through investigation of CDKN2A variants through candidate gene approach as well as by genome wide association studies (GWAS). Methods In the present study we genotyped, 25 SNPs that tag 273 variants on chromosome 9p21 in 837 melanoma cases and 1154 controls from Spain. Ten SNPs were selected based on previous associations, reported in GWAS, with either melanocytic nevi or melanoma risk or both. The other 15 SNPs were selected to fine map the CDKN2A gene region. Results All the 10 variants selected from the GWAS showed statistically significant association with melanoma risk. Statistically significant association with melanoma risk was also observed for the carriers of the variant T-allele of rs3088440 (540 C>T) at the 3’ UTR of CDKN2A gene with an OR 1.52 (95% CI 1.14-2.04). Interaction analysis between risk associated polymorphisms and previously genotyped MC1R variants, in the present study, did not show any statistically significant association. Statistical significant association was observed for the interaction between phototypes and the rs10811629 (located in intron 5 of MTAP). The strongest association was observed between the homozygous carrier of the A–allele and phototype II with an OR of 15.93 (95% CI 5.34-47.54). Conclusions Our data confirmed the association of different variants at chromosome 9p21 with melanoma risk and we also found an association of a variant with skin phototypes.
Collapse
Affiliation(s)
- Livia Maccioni
- Division of Molecular Genetic Epidemiology, German Cancer Research Centre (DKFZ), Im Neuenheimer Feld 580, D-69120, Heidelberg, Germany
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Reissmann M, Ludwig A. Pleiotropic effects of coat colour-associated mutations in humans, mice and other mammals. Semin Cell Dev Biol 2013; 24:576-86. [PMID: 23583561 DOI: 10.1016/j.semcdb.2013.03.014] [Citation(s) in RCA: 106] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 03/27/2013] [Accepted: 03/28/2013] [Indexed: 12/20/2022]
Abstract
The characterisation of the pleiotropic effects of coat colour-associated mutations in mammals illustrates that sensory organs and nerves are particularly affected by disorders because of the shared origin of melanocytes and neurocytes in the neural crest; e.g. the eye-colour is a valuable indicator of disorders in pigment production and eye dysfunctions. Disorders related to coat colour-associated alleles also occur in the skin (melanoma), reproductive tract and immune system. Additionally, the coat colour phenotype of an individual influences its general behaviour and fitness. Mutations in the same genes often produce similar coat colours and pleiotropic effects in different species (e.g., KIT [reproductive disorders, lethality], EDNRB [megacolon] and LYST [CHS]). Whereas similar disorders and similar-looking coat colour phenotypes sometimes have a different genetic background (e.g., deafness [EDN3/EDNRB, MITF, PAX and SNAI2] and visual diseases [OCA2, RAB38, SLC24A5, SLC45A2, TRPM1 and TYR]). The human predilection for fancy phenotypes that ignore disorders and genetic defects is a major driving force for the increase of pleiotropic effects in domestic species and laboratory subjects since domestication has commenced approximately 18,000 years ago.
Collapse
Key Words
- AS
- ASIP
- ATRN
- Agouti signalling protein
- Albino
- Angelman syndrome
- Attractin (mahogany)
- BLOC
- Biogenesis of lysosomal organelles complex
- CCSD
- CHS
- CSD
- CSNB
- Canine congenital sensorineural deafness
- Chediak-Higashi syndrome
- Coat colour gene
- Congenital sensorineural deafness
- Congenital stationary night blindness
- Disorder
- EDN3
- EDNRB
- Endothelin 3
- Endothelin receptor type B
- Epistasis
- Fitness
- GS
- Griscelli syndrome (type 1 or 2)
- HPS
- HSCR
- Hermansky-Pudlak syndrome with different types
- Hirschsprung disease
- IPE
- Iris pigment epithelium
- KIT
- KIT ligand (steel factor)
- KITLG
- LFS
- LYST
- Lavender foal syndrome
- Lethal
- Leucism
- Lysosomal trafficking regulator
- MC1R
- MCOA
- MCOLN3
- MGRN1
- MITF
- MYO5A
- Mahogunin ring finger 1 (E3 ubiquitin protein ligase)
- Melanocortin 1 receptor
- Melanoma
- Microphthalmia-associated transcription factor
- Mucolipin 3 (TRPML3)
- Multiple congenital ocular anomalies
- Myosin VA (heavy chain 12, myoxin)
- OA
- OCA
- OCA2
- OLWS
- OSTM1
- Ocular albinism
- Oculocutaneous albinism II (pink-eye dilution homolog)
- Oculocutaneous albinism type 1–4
- Osteopetrosis associated transmembrane protein 1 (Grey lethal osteopetrosis)
- Overo lethal white syndrome
- PAX3
- PMEL
- PWS
- Paired box 3
- Pleiotropy
- Prader-Willi syndrome
- Premelanosome protein (Pmel17, SILV)
- RAB27A
- RAB27A member RAS oncogene family
- RAB38
- RAB38 member RAS oncogene family
- RPE
- Reproduction
- Retinal pigmented epithelium
- SLC24A5
- SLC2A9
- SLC45A2
- SNAI2
- STX17
- Snail homolog 2 (Drosophila), (SLUG), SOX10, SRY (sex determining region Y)-box 10
- Solute carrier family 2 (facilitated glucose transporter), member 9
- Solute carrier family 24, member 5
- Solute carrier family 45, member 2, MATP
- Syntaxin 17
- TRPM1
- TYR
- Tameness
- Transient receptor potential cation channel, subfamily M, member 1 (melastatin-1)
- Tyrosinase, TYRP1, Tyrosinase-related protein 1
- V-kit Hardy-Zuckerman 4 feline sarcoma viral oncogene homolog, tyrosine kinase receptor (c-kit)
- WS
- Waardenburg syndrome (type 1, type 2 combined with Tietz syndrome type 3 Klein-Waardenburg syndrome, type 4 Waardenburg-Shah syndrome)
- alpha-melanocyte-stimulating hormone
- αMSH
Collapse
Affiliation(s)
- Monika Reissmann
- Humboldt University Berlin, Department for Crop and Animal Sciences, Berlin, Germany.
| | | |
Collapse
|