Network


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

Hotspot


Dive into the research topics where Mark Harland is active.

Publication


Featured researches published by Mark Harland.


Nature Genetics | 2009

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

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

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


Cancer Research | 2006

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

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

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


Nature | 2011

A novel recurrent mutation in MITF predisposes to familial and sporadic melanoma

Satoru Yokoyama; Susan L. Woods; Glen M. Boyle; Lauren G. Aoude; Stuart Macgregor; Victoria Zismann; Michael Gartside; Anne E. Cust; Rizwan Haq; Mark Harland; John C. Taylor; David L. Duffy; Kelly Holohan; Ken Dutton-Regester; Jane M. Palmer; Vanessa F. Bonazzi; Mitchell S. Stark; Judith Symmons; Matthew H. Law; Christopher W. Schmidt; Cathy Lanagan; Linda O’Connor; Elizabeth A. Holland; Helen Schmid; Judith A. Maskiell; Jodie Jetann; Megan Ferguson; Mark A. Jenkins; Richard F. Kefford; Graham G. Giles

So far, two genes associated with familial melanoma have been identified, accounting for a minority of genetic risk in families. Mutations in CDKN2A account for approximately 40% of familial cases, and predisposing mutations in CDK4 have been reported in a very small number of melanoma kindreds. Here we report the whole-genome sequencing of probands from several melanoma families, which we performed in order to identify other genes associated with familial melanoma. We identify one individual carrying a novel germline variant (coding DNA sequence c.G1075A; protein sequence p.E318K; rs149617956) in the melanoma-lineage-specific oncogene microphthalmia-associated transcription factor (MITF). Although the variant co-segregated with melanoma in some but not all cases in the family, linkage analysis of 31 families subsequently identified to carry the variant generated a log of odds (lod) score of 2.7 under a dominant model, indicating E318K as a possible intermediate risk variant. Consistent with this, the E318K variant was significantly associated with melanoma in a large Australian case–control sample. Likewise, it was similarly associated in an independent case–control sample from the United Kingdom. In the Australian sample, the variant allele was significantly over-represented in cases with a family history of melanoma, multiple primary melanomas, or both. The variant allele was also associated with increased naevus count and non-blue eye colour. Functional analysis of E318K showed that MITF encoded by the variant allele had impaired sumoylation and differentially regulated several MITF targets. These data indicate that MITF is a melanoma-predisposition gene and highlight the utility of whole-genome sequencing to identify novel rare variants associated with disease susceptibility.


Journal of Medical Genetics | 2006

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

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

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


Nature Genetics | 2011

Genome-wide association study identifies three new melanoma susceptibility loci

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

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


Nature Genetics | 2014

POT1 loss-of-function variants predispose to familial melanoma

Carla Daniela Robles-Espinoza; Mark Harland; Andrew J. Ramsay; Lauren G. Aoude; Zhihao Ding; Karen A. Pooley; Antonia L. Pritchard; Jessamy Tiffen; Mia Petljak; Jane M. Palmer; Judith Symmons; Peter Johansson; Mitchell S. Stark; Michael Gartside; Helen Snowden; Grant W. Montgomery; Nicholas G. Martin; Jimmy Z. Liu; Jiyeon Choi; Matthew Makowski; Kevin M. Brown; Alison M. Dunning; Thomas M. Keane; Carlos López-Otín; Nelleke A. Gruis; Nicholas K. Hayward; D. Timothy Bishop; Julia Newton-Bishop; David J. Adams

Deleterious germline variants in CDKN2A account for around 40% of familial melanoma cases, and rare variants in CDK4, BRCA2, BAP1 and the promoter of TERT have also been linked to the disease. Here we set out to identify new high-penetrance susceptibility genes by sequencing 184 melanoma cases from 105 pedigrees recruited in the UK, The Netherlands and Australia that were negative for variants in known predisposition genes. We identified families where melanoma cosegregates with loss-of-function variants in the protection of telomeres 1 gene (POT1), with a proportion of family members presenting with an early age of onset and multiple primary tumors. We show that these variants either affect POT1 mRNA splicing or alter key residues in the highly conserved oligonucleotide/oligosaccharide-binding (OB) domains of POT1, disrupting protein-telomere binding and leading to increased telomere length. These findings suggest that POT1 variants predispose to melanoma formation via a direct effect on telomeres.


Molecular and Cellular Biology | 2002

Biallelic mutations in p16(INK4a) confer resistance to Ras- and Ets-induced senescence in human diploid fibroblasts.

Thomas J. G. Huot; Janice Rowe; Mark Harland; Sarah Drayton; Sharon M. Brookes; Chandra Gooptu; Patricia Purkis; Michael Fried; Veronique Bataille; Eiji Hara; Julia Newton-Bishop; Gordon Peters

ABSTRACT The INK4a/ARF tumor suppressor locus is implicated in the senescence-like growth arrest provoked by oncogenic Ras in primary cells. INK4a and ARF are distinct proteins encoded by transcripts in which a shared exon is decoded in alternative reading frames. Here we analyze dermal fibroblasts (designated Q34) from an individual carrying independent missense mutations in each copy of the common exon. Both mutations alter the amino acid sequence of INK4a and functionally impair the protein, although they do so to different degrees. Only one of the mutations affects the sequence of ARF, causing an apparently innocuous change near its carboxy terminus. Unlike normal human fibroblasts, Q34 cells are not permanently arrested by Ras or its downstream effectors Ets1 and Ets2. Moreover, ectopic Ras enables the cells to grow as anchorage-independent colonies, and in relatively young Q34 cells anchorage independence can be achieved without addition of telomerase or perturbation of the p53 pathway. Whereas ARF plays the principal role in Ras-induced arrest of mouse fibroblasts, our data imply that INK4a assumes this role in human fibroblasts.


Genes, Chromosomes and Cancer | 2005

A large Norwegian family with inherited malignant melanoma, multiple atypical nevi, and CDK4 mutation.

Magne B. Grimstvedt; Solrun J. Steine; Mark Harland; Marie-Françoise Avril; Nicholas K. Hayward; Lars A. Akslen

Mutations in two loci encoding cell‐cycle‐regulatory proteins have been shown to cause familial malignant melanoma. About 20% of melanoma‐prone families bear a mutation in the CDKN2A locus, which encodes two unrelated proteins, p16INK4A and p14ARF. Mutations in the other locus, CDK4, are much rarer and have been linked to the disease in only three families worldwide. In the 1960s, a large Norwegian pedigree with multiple atypical nevi and malignant melanomas was identified. Subsequently, six generations and more than 100 family members were traced and 20 cases of melanoma verified. In this article, we report that CDK4 codon 24 is mutated from CGT to CAT (Arg24His) in this unusually large melanoma kindred. Intriguingly, one of the family members had ocular melanoma, but the CDK4 mutation could not be detected in archival tissue samples from this subject. Thus, the case of ocular melanoma in this family was sporadic, suggesting an etiology different from that of the skin tumors. The CDK4 mutation in the Norwegian family was identical to that in melanoma families in France, Australia, and England. Haplotype analysis using microsatellite markers flanking the CDK4 gene and single‐nucleotide polymorphisms within the gene did not support the possibility that there was a common founder, but rather indicated at least two independent mutational events. All CDK4 melanoma families known to date have a substitution of amino acid 24. In addition to resulting from selection pressure, this observation may be explained by the CG dinucleotide of codon 24 representing a mutational hot spot in the CDK4 gene.


Nature Genetics | 2015

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

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

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


Genes, Chromosomes and Cancer | 2000

Mutation screening of the CDKN2A promoter in melanoma families

Mark Harland; Elizabeth A. Holland; Paola Ghiorzo; Michela Mantelli; Giovanna Bianchi-Scarrà; Alisa M. Goldstein; Margaret A. Tucker; Bruce A.J. Ponder; Graham J. Mann; D. Timothy Bishop; Julia A. Newton Bishop

Germline mutations of CDKN2A, at 9p21, are responsible for predisposition to melanoma in some families. However, evidence of linkage to 9p21 has been demonstrated in a significant proportion of kindreds with no detectable mutations in CDKN2A. It is possible that mutations in noncoding regions may be responsible for predisposition to melanoma in these families. We have analyzed approximately 1 kb of the CDKN2A promoter upstream of the start codon in an attempt to identify causal mutations in 107 melanoma families. Four sequence variants were detected. Two of these (A‐191G and A‐493T) did not segregate with disease and were present in a control population at a comparable frequency, indicating that they are unlikely to predispose to melanoma. The A‐493T variant appeared to be in linkage disequilibrium with the previously described CDKN2A polymorphism Ala148Thr. The variant G‐735A was detected in the control population, but segregation of this variant with melanoma within families could not be discounted. The fourth variant (G‐34T), located in the 5′ UTR, creates an aberrant initiation codon. This variant appeared to segregate with melanoma and was not detected in a control population. G‐34T has recently been identified in a subset of Canadian melanoma families and was concluded to be associated with predisposition to melanoma. The creation of an aberrant initiation site in the 5′ UTR may have an important role in carcinogenesis in a small percentage of families; however, mutations in the CDKN2A promoter appear to have a limited role in predisposition to melanoma. Genes Chromosomes Cancer 28:45–57, 2000.

Collaboration


Dive into the Mark Harland's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Nicholas K. Hayward

QIMR Berghofer Medical Research Institute

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge