Network


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

Hotspot


Dive into the research topics where Giulia Orlando is active.

Publication


Featured researches published by Giulia Orlando.


Nature Genetics | 2017

Identification of 19 new risk loci and potential regulatory mechanisms influencing susceptibility to testicular germ cell tumor

Kevin Litchfield; Max Levy; Giulia Orlando; Chey Loveday; Philip J. Law; Gabriele Migliorini; Amy Holroyd; Peter Broderick; Robert Karlsson; Trine B. Haugen; Wenche Kristiansen; Jérémie Nsengimana; Kerry Fenwick; Ioannis Assiotis; Zsofia Kote-Jarai; Alison M. Dunning; Kenneth Muir; Julian Peto; Rosalind Eeles; Douglas F. Easton; Darshna Dudakia; Nick Orr; Nora Pashayan; D. Timothy Bishop; Alison Reid; Robert Huddart; Janet Shipley; Tom Grotmol; Fredrik Wiklund; Richard S. Houlston

Genome-wide association studies (GWAS) have transformed understanding of susceptibility to testicular germ cell tumors (TGCTs), but much of the heritability remains unexplained. Here we report a new GWAS, a meta-analysis with previous GWAS and a replication series, totaling 7,319 TGCT cases and 23,082 controls. We identify 19 new TGCT risk loci, roughly doubling the number of known TGCT risk loci to 44. By performing in situ Hi-C in TGCT cells, we provide evidence for a network of physical interactions among all 44 TGCT risk SNPs and candidate causal genes. Our findings implicate widespread disruption of developmental transcriptional regulators as a basis of TGCT susceptibility, consistent with failed primordial germ cell differentiation as an initiating step in oncogenesis. Defective microtubule assembly and dysregulation of KIT–MAPK signaling also feature as recurrently disrupted pathways. Our findings support a polygenic model of risk and provide insight into the biological basis of TGCT.


Scientific Reports | 2017

Genome-wide association analysis of chronic lymphocytic leukaemia, Hodgkin lymphoma and multiple myeloma identifies pleiotropic risk loci

Philip J. Law; Amit Sud; Jonathan S. Mitchell; Marc Henrion; Giulia Orlando; Oleg Lenive; Peter Broderick; Helen E. Speedy; David C. Johnson; Martin Kaiser; Niels Weinhold; Rosie Cooke; Nicola J. Sunter; Graham Jackson; Geoffrey Summerfield; Robert J. Harris; Andrew R. Pettitt; David Allsup; Jonathan Carmichael; James R Bailey; Guy Pratt; Thahira Rahman; Chris Pepper; Christopher Fegan; Elke Pogge von Strandmann; Andreas Engert; Asta Försti; Bowang Chen; Miguel Inacio da Silva Filho; Hauke Thomsen

B-cell malignancies (BCM) originate from the same cell of origin, but at different maturation stages and have distinct clinical phenotypes. Although genetic risk variants for individual BCMs have been identified, an agnostic, genome-wide search for shared genetic susceptibility has not been performed. We explored genome-wide association studies of chronic lymphocytic leukaemia (CLL, N = 1,842), Hodgkin lymphoma (HL, N = 1,465) and multiple myeloma (MM, N = 3,790). We identified a novel pleiotropic risk locus at 3q22.2 (NCK1, rs11715604, P = 1.60 × 10−9) with opposing effects between CLL (P = 1.97 × 10−8) and HL (P = 3.31 × 10−3). Eight established non-HLA risk loci showed pleiotropic associations. Within the HLA region, Ser37 + Phe37 in HLA-DRB1 (P = 1.84 × 10−12) was associated with increased CLL and HL risk (P = 4.68 × 10−12), and reduced MM risk (P = 1.12 × 10−2), and Gly70 in HLA-DQB1 (P = 3.15 × 10−10) showed opposing effects between CLL (P = 3.52 × 10−3) and HL (P = 3.41 × 10−9). By integrating eQTL, Hi-C and ChIP-seq data, we show that the pleiotropic risk loci are enriched for B-cell regulatory elements, as well as an over-representation of binding of key B-cell transcription factors. These data identify shared biological pathways influencing the development of CLL, HL and MM. The identification of these risk loci furthers our understanding of the aetiological basis of BCMs.


Nature Communications | 2016

Multiple myeloma risk variant at 7p15.3 creates an IRF4-binding site and interferes with CDCA7L expression

Ni Li; David C. Johnson; Niels Weinhold; James B. Studd; Giulia Orlando; Fabio Mirabella; Jonathan S. Mitchell; Tobias Meissner; Martin Kaiser; Hartmut Goldschmidt; Kari Hemminki; Gareth J. Morgan; Richard S. Houlston

Genome-wide association studies have identified several risk loci for multiple myeloma (MM); however, the mechanisms by which they influence MM are unknown. Here by using genetic association data and functional characterization, we demonstrate that rs4487645 G>T, the most highly associated variant (P = 5.30 × 10−25), resides in an enhancer element 47 kb upstream of the transcription start site of c-Myc-interacting CDCA7L. The G-risk allele, associated with increased CDCA7L expression (P=1.95 × 10−36), increases IRF4 binding and the enhancer interacts with the CDCA7L promoter. We show that suppression of CDCA7L limits MM proliferation through apoptosis, and increased CDCA7L expression is associated with adverse patient survival. These findings implicate IRF4-mediated CDCA7L expression in MM biology and indicate how germline variation might confer susceptibility to MM.


540-546 | 2018

Genome-wide association study and meta-analysis in Northern European populations replicate multiple colorectal cancer risk loci

Tomas Tanskanen; Linda van den Berg; Niko Välimäki; Mervi Aavikko; Eivind Ness-Jensen; Kristian Hveem; Yvonne Wettergren; Elinor Bexe Lindskog; Neeme Tõnisson; Andres Metspalu; Kaisa Silander; Giulia Orlando; Philip J. Law; Sari Tuupanen; Alexandra E. Gylfe; Ulrika A. Hänninen; Tatiana Cajuso; Johanna Kondelin; Antti-Pekka Sarin; Eero Pukkala; Pekka Jousilahti; Veikko Salomaa; Samuli Ripatti; Aarno Palotie; Heikki Järvinen; Laura Renkonen-Sinisalo; Anna Lepistö; Jan Böhm; Jukka-Pekka Mecklin; Nada A. Al-Tassan

Genome‐wide association studies have been successful in elucidating the genetic basis of colorectal cancer (CRC), but there remains unexplained variability in genetic risk. To identify new risk variants and to confirm reported associations, we conducted a genome‐wide association study in 1,701 CRC cases and 14,082 cancer‐free controls from the Finnish population. A total of 9,068,015 genetic variants were imputed and tested, and 30 promising variants were studied in additional 11,647 cases and 12,356 controls of European ancestry. The previously reported association between the single‐nucleotide polymorphism (SNP) rs992157 (2q35) and CRC was independently replicated (p = 2.08 × 10−4; OR, 1.14; 95% CI, 1.06–1.23), and it was genome‐wide significant in combined analysis (p = 1.50 × 10−9; OR, 1.12; 95% CI, 1.08–1.16). Variants at 2q35, 6p21.2, 8q23.3, 8q24.21, 10q22.3, 10q24.2, 11q13.4, 11q23.1, 14q22.2, 15q13.3, 18q21.1, 20p12.3 and 20q13.33 were associated with CRC in the Finnish population (false discovery rate < 0.1), but new risk loci were not found. These results replicate the effects of multiple loci on the risk of CRC and identify shared risk alleles between the Finnish population isolate and outbred populations.


Current protocols in human genetics | 2018

Capture Hi‐C Library Generation and Analysis to Detect Chromatin Interactions

Giulia Orlando; Ben Kinnersley; Richard S. Houlston

Chromosome conformation capture (3C), coupled with next‐generation sequencing (Hi‐C), provides a means for deciphering not only the principles underlying genome folding and architecture, but more broadly, the role 3D chromatin structure plays in gene regulation and the replication and repair of DNA. The recently implemented modification, in situ Hi‐C, maintains nuclear integrity during digestion and ligation steps, reducing random ligation of Hi‐C fragments. Although Hi‐C allows for genome‐wide characterization of chromatin contacts, it requires high‐depth sequencing to discover significant contacts. To address this, Capture Hi‐C (CHi‐C) enriches standard Hi‐C libraries for regions of biological interest, for example by specifically targeting gene promoters, aiding identification of biologically significant chromatin interactions compared to conventional Hi‐C, for an equivalent number of sequence reads. Illustrating the application of CHi‐C applied to genome‐wide analysis of chromatin interactions with promoters, we detail the protocols for in situ Hi‐C and CHi‐C library generation for sequencing, as well as the bioinformatics tools for data analysis.


Cell Reports | 2017

Genetic Predisposition to Multiple Myeloma at 5q15 Is Mediated by an ELL2 Enhancer Polymorphism

Ni L. Li; David C. Johnson; Niels Weinhold; Scott Kimber; Sara E. Dobbins; Jonathan S. Mitchell; Ben Kinnersley; Amit Sud; Philip J. Law; Giulia Orlando; Matthew Scales; Christopher P. Wardell; Asta Försti; Phuc H. Hoang; Molly Went; Amy Holroyd; Fadi Hariri; Tomi Pastinen; Tobias Meissner; Hartmut Goldschmidt; Kari Hemminki; Gareth J. Morgan; Martin Kaiser; Richard S. Houlston

Summary Multiple myeloma (MM) is a malignancy of plasma cells. Genome-wide association studies have shown that variation at 5q15 influences MM risk. Here, we have sought to decipher the causal variant at 5q15 and the mechanism by which it influences tumorigenesis. We show that rs6877329 G > C resides in a predicted enhancer element that physically interacts with the transcription start site of ELL2. The rs6877329-C risk allele is associated with reduced enhancer activity and lowered ELL2 expression. Since ELL2 is critical to the B cell differentiation process, reduced ELL2 expression is consistent with inherited genetic variation contributing to arrest of plasma cell development, facilitating MM clonal expansion. These data provide evidence for a biological mechanism underlying a hereditary risk of MM at 5q15.


Nature Genetics | 2018

Promoter capture Hi-C-based identification of recurrent noncoding mutations in colorectal cancer

Giulia Orlando; Philip J. Law; Alex J. Cornish; Sara E. Dobbins; Daniel Chubb; Peter Broderick; Kevin Litchfield; Fadi Hariri; Tomi Pastinen; Cameron S. Osborne; Jussi Taipale; Richard S. Houlston

Efforts are being directed to systematically analyze the non-coding regions of the genome for cancer-driving mutations1–6. cis-regulatory elements (CREs) represent a highly enriched subset of the non-coding regions of the genome in which to search for such mutations. Here we use high-throughput chromosome conformation capture techniques (Hi-C) for 19,023 promoter fragments to catalog the regulatory landscape of colorectal cancer in cell lines, mapping CREs and integrating these with whole-genome sequence and expression data from The Cancer Genome Atlas7,8. We identify a recurrently mutated CRE interacting with the ETV1 promoter affecting gene expression. ETV1 expression influences cell viability and is associated with patient survival. We further refine our understanding of the regulatory effects of copy-number variations, showing that RASL11A is targeted by a previously identified enhancer amplification1. This study reveals new insights into the complex genetic alterations driving tumor development, providing a paradigm for employing chromosome conformation capture to decipher non-coding CREs relevant to cancer biology.Promoter capture Hi-C in colorectal cancer cells integrated with cancer genome and expression data identifies a noncoding, cis-regulatory element that is recurrently mutated in cancer, affecting ETV1 expression, cell viability and patient survival.


Blood | 2018

Genome-wide association study implicates immune dysfunction in the development of Hodgkin lymphoma.

Amit Sud; Hauke Thomsen; Giulia Orlando; Asta Försti; Philip J. Law; Peter Broderick; Rosie Cooke; Fadi Hariri; Tomi Pastinen; Douglas F. Easton; Paul Pharoah; Alison M. Dunning; Julian Peto; Federico Canzian; Rosalind Eeles; Zsofia Kote-Jarai; Kenneth Muir; Nora Pashayan; Daniele Campa; Per Hoffmann; Markus M. Nöthen; Karl-Heinz Jöckel; Elke Pogge von Strandmann; Anthony J. Swerdlow; Andreas Engert; Nick Orr; Kari Hemminki; Richard S. Houlston

To further our understanding of inherited susceptibility to Hodgkin lymphoma (HL), we performed a meta-analysis of 7 genome-wide association studies totaling 5325 HL cases and 22 423 control patients. We identify 5 new HL risk loci at 6p21.31 (rs649775; P = 2.11 × 10-10), 6q23.3 (rs1002658; P = 2.97 × 10-8), 11q23.1 (rs7111520; P = 1.44 × 10-11), 16p11.2 (rs6565176; P = 4.00 × 10-8), and 20q13.12 (rs2425752; P = 2.01 × 10-8). Integration of gene expression, histone modification, and in situ promoter capture Hi-C data at the 5 new and 13 known risk loci implicates dysfunction of the germinal center reaction, disrupted T-cell differentiation and function, and constitutive NF-κB activation as mechanisms of predisposition. These data provide further insights into the genetic susceptibility and biology of HL.


Nature Communications | 2017

Genome-wide association study of classical Hodgkin lymphoma identifies key regulators of disease susceptibility

Amit Sud; Hauke Thomsen; Philip J. Law; Asta Försti; Miguel Inacio da Silva Filho; Amy Holroyd; Peter Broderick; Giulia Orlando; Oleg Lenive; Lauren B. Wright; Rosie Cooke; Douglas F. Easton; Paul Pharoah; Alison M. Dunning; Julian Peto; Federico Canzian; Rosalind Eeles; Zsofia Kote-Jarai; Kenneth W. Muir; Nora Pashayan; Per Hoffmann; Markus M. Nöthen; Karl-Heinz Jöckel; Elke Pogge von Strandmann; Tracy Lightfoot; Eleanor Kane; Eve Roman; Annette Lake; Dorothy Montgomery; Ruth F. Jarrett

Several susceptibility loci for classical Hodgkin lymphoma have been reported. However, much of the heritable risk is unknown. Here, we perform a meta-analysis of two existing genome-wide association studies, a new genome-wide association study, and replication totalling 5,314 cases and 16,749 controls. We identify risk loci for all classical Hodgkin lymphoma at 6q22.33 (rs9482849, P = 1.52 × 10−8) and for nodular sclerosis Hodgkin lymphoma at 3q28 (rs4459895, P = 9.43 × 10−17), 6q23.3 (rs6928977, P = 4.62 × 10−11), 10p14 (rs3781093, P = 9.49 × 10−13), 13q34 (rs112998813, P = 4.58 × 10−8) and 16p13.13 (rs34972832, P = 2.12 × 10−8). Additionally, independent loci within the HLA region are observed for nodular sclerosis Hodgkin lymphoma (rs9269081, HLA-DPB1*03:01, Val86 in HLA-DRB1) and mixed cellularity Hodgkin lymphoma (rs1633096, rs13196329, Val86 in HLA-DRB1). The new and established risk loci localise to areas of active chromatin and show an over-representation of transcription factor binding for determinants of B-cell development and immune response.Classical Hodgkin lymphoma is a cancer that originates in lymph nodes. Little is known about its genetic susceptibility. Here, the authors combined existing and new genome-wide association studies to identify risk loci for classical Hodgkin lymphoma at 6q22.33, and nodular sclerosis Hodgkin lymphoma at 3q28, 6q23.3, 10p14, 13q34, 16p13.13.


WOS | 2016

Variation at 2q35 (PNKD and TMBIM1) influences colorectal cancer risk and identifies a pleiotropic effect with inflammatory bowel disease

Giulia Orlando; Philip J. Law; Kimmo Palin; Sari Tuupanen; Alexandra E. Gylfe; Ulrika A. Hänninen; Tatiana Cajuso; Tomas Tanskanen; Johanna Kondelin; Eevi Kaasinen; Antti-Pekka Sarin; Jaakko Kaprio; Johan G. Eriksson; Harri Rissanen; Paul Knekt; Eero Pukkala; Pekka Jousilahti; Veikko Salomaa; Samuli Ripatti; Aarno Palotie; Heikki Järvinen; Laura Renkonen-Sinisalo; Anna Lepistö; Jan Böhm; Jukka-Pekka Mecklin; Nada A. Al-Tassan; Claire Palles; Lynn Martin; Ella Barclay; Albert Tenesa

Collaboration


Dive into the Giulia Orlando's collaboration.

Top Co-Authors

Avatar

Philip J. Law

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Richard S. Houlston

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Peter Broderick

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar

Amit Sud

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Amy Holroyd

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jonathan S. Mitchell

Institute of Cancer Research

View shared research outputs
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge