Network


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

Hotspot


Dive into the research topics where Espen Melum is active.

Publication


Featured researches published by Espen Melum.


Gastroenterology | 2010

Genome-Wide Association Analysis in Primary Sclerosing Cholangitis

Tom H. Karlsen; Andre Franke; Espen Melum; Arthur Kaser; Johannes R. Hov; Tobias Balschun; Benedicte A. Lie; Annika Bergquist; Christoph Schramm; Tobias J. Weismüller; Daniel Gotthardt; Christian Rust; Eva Philipp; Teresa Fritz; Liesbet Henckaerts; Rinse K. Weersma; Pieter Stokkers; Cyriel Y. Ponsioen; Cisca Wijmenga; Martina Sterneck; Michael Nothnagel; Jochen Hampe; Andreas Teufel; Heiko Runz; Philip Rosenstiel; Adolf Stiehl; Severine Vermeire; Ulrich Beuers; Michael P. Manns; Erik Schrumpf

BACKGROUND & AIMS We aimed to characterize the genetic susceptibility to primary sclerosing cholangitis (PSC) by means of a genome-wide association analysis of single nucleotide polymorphism (SNP) markers. METHODS A total of 443,816 SNPs on the Affymetrix SNP Array 5.0 (Affymetrix, Santa Clara, CA) were genotyped in 285 Norwegian PSC patients and 298 healthy controls. Associations detected in this discovery panel were re-examined in independent case-control panels from Scandinavia (137 PSC cases and 368 controls), Belgium/The Netherlands (229 PSC cases and 735 controls), and Germany (400 cases and 1832 controls). RESULTS The strongest associations were detected near HLA-B at chromosome 6p21 (rs3099844: odds ratio [OR], 4.8; 95% confidence interval [CI], 3.6-6.5; P = 2.6 x 10(-26); and rs2844559: OR, 4.7; 95% CI, 3.5-6.4; P = 4.2 x 10(-26) in the discovery panel). Outside the HLA complex, rs9524260 at chromosome 13q31 showed significant associations in 3 of 4 study panels. Lentiviral silencing of glypican 6, encoded at this locus, led to the up-regulation of proinflammatory markers in a cholangiocyte cell line. Of 15 established ulcerative colitis susceptibility loci, significant replication was obtained at chromosomes 2q35 and 3p21 (rs12612347: OR, 1.26; 95% CI, 1.06-1.50; and rs3197999: OR, 1.22; 95% CI, 1.02-1.47, respectively), with circumstantial evidence supporting the G-protein-coupled bile acid receptor 1 and macrophage-stimulating 1, respectively, as the likely disease genes. CONCLUSIONS Strong HLA associations and a subset of genes involved in bile homeostasis and other inflammatory conditions constitute key components of the genetic architecture of PSC.


Nature Genetics | 2011

Genome-wide association analysis in primary sclerosing cholangitis identifies two non-HLA susceptibility loci.

Espen Melum; Andre Franke; Christoph Schramm; Tobias J. Weismüller; Daniel Gotthardt; Felix Offner; Brian D. Juran; Jon K. Laerdahl; Verena Labi; Einar Björnsson; Rinse K. Weersma; Liesbet Henckaerts; Andreas Teufel; Christian Rust; Eva Ellinghaus; Tobias Balschun; Kirsten Muri Boberg; David Ellinghaus; Annika Bergquist; Peter Sauer; Euijung Ryu; Johannes R. Hov; Jochen Wedemeyer; Björn Lindkvist; Michael Wittig; Robert J. Porte; Kristian Holm; Christian Gieger; H-Erich Wichmann; Pieter Stokkers

Primary sclerosing cholangitis (PSC) is a chronic bile duct disease affecting 2.4–7.5% of individuals with inflammatory bowel disease. We performed a genome-wide association analysis of 2,466,182 SNPs in 715 individuals with PSC and 2,962 controls, followed by replication in 1,025 PSC cases and 2,174 controls. We detected non-HLA associations at rs3197999 in MST1 and rs6720394 near BCL2L11 (combined P = 1.1 × 10−16 and P = 4.1 × 10−8, respectively).


Journal of Hepatology | 2012

Extended analysis of a genome-wide association study in primary sclerosing cholangitis detects multiple novel risk loci

Trine Folseraas; Espen Melum; Philipp Rausch; Brian D. Juran; Eva Ellinghaus; Alexey Shiryaev; Jon K. Laerdahl; David Ellinghaus; Christoph Schramm; Tobias J. Weismüller; Daniel Gotthardt; Johannes R. Hov; O. P. F. Clausen; Rinse K. Weersma; Marcel Janse; Kirsten Muri Boberg; Einar Björnsson; Hanns-Ulrich Marschall; Isabelle Cleynen; Philip Rosenstiel; Kristian Holm; Andreas Teufel; Christian Rust; Christian Gieger; H-Erich Wichmann; Annika Bergquist; Euijung Ryu; Cyriel Y. Ponsioen; Heiko Runz; Martina Sterneck

BACKGROUND & AIMS A limited number of genetic risk factors have been reported in primary sclerosing cholangitis (PSC). To discover further genetic susceptibility factors for PSC, we followed up on a second tier of single nucleotide polymorphisms (SNPs) from a genome-wide association study (GWAS). METHODS We analyzed 45 SNPs in 1221 PSC cases and 3508 controls. The association results from the replication analysis and the original GWAS (715 PSC cases and 2962 controls) were combined in a meta-analysis comprising 1936 PSC cases and 6470 controls. We performed an analysis of bile microbial community composition in 39 PSC patients by 16S rRNA sequencing. RESULTS Seventeen SNPs representing 12 distinct genetic loci achieved nominal significance (p(replication) <0.05) in the replication. The most robust novel association was detected at chromosome 1p36 (rs3748816; p(combined)=2.1 × 10(-8)) where the MMEL1 and TNFRSF14 genes represent potential disease genes. Eight additional novel loci showed suggestive evidence of association (p(repl) <0.05). FUT2 at chromosome 19q13 (rs602662; p(comb)=1.9 × 10(-6), rs281377; p(comb)=2.1 × 10(-6) and rs601338; p(comb)=2.7 × 10(-6)) is notable due to its implication in altered susceptibility to infectious agents. We found that FUT2 secretor status and genotype defined by rs601338 significantly influence biliary microbial community composition in PSC patients. CONCLUSIONS We identify multiple new PSC risk loci by extended analysis of a PSC GWAS. FUT2 genotype needs to be taken into account when assessing the influence of microbiota on biliary pathology in PSC.


Hepatology | 2011

IL28B genetic variation and treatment response in patients with hepatitis C virus genotype 3 infection

Amir Moghaddam; Espen Melum; Nils Reinton; Helmer Ring-Larsen; Hans Verbaan; Kristian Bjøro; Olav Dalgard

Polymorphisms near the IL28B gene, which code for interferon (IFN)‐λ3, predict response to pegylated interferon‐α (PEG‐IFN) and ribavirin treatment in hepatitis C virus (HCV) genotype 1 infected patients. Follow‐up studies of the effect of IL28B gene in HCV non–genotype 1 infected patients have almost always used predominantly HCV genotype 2–infected or mixed genotype 2/3–infected cohorts with results partly conflicting with HCV genotype 1. We performed a retrospective analysis of 281 patients infected with HCV genotype 3 for association of response to therapy with IL28B polymorphisms. We found that the HCV genotype 1 responder genotypes at rs12979860 and rs8099917 did not associate with sustained virological response to PEG‐IFN/ribavirin therapy. However, the responder genotypes of both SNPs showed association with rapid viral response measured at 4 weeks (rs12979860, P = 3 × 10−5; rs8099917, P = 3 × 10−4). In multivariate analysis, age (<40 years), baseline viral load (<4 × 105 IU/mL) and the responder genotypes of SNPs rs12979860 or rs8099917 remained significant independent predictors of rapid viral response to therapy. Furthermore, we show that IL28B polymorphisms are associated with relapse in patients who achieve rapid viral response to PEG‐IFN/ribavirin therapy. The responder genotypes also showed association with markers of stage and activity of liver disease, namely high aspartate aminotransferase platelet ratio index (APRI, rs12979860, P = 0.018; rs8099917, not significant) and high alanine aminotransferase (ALT, rs12979860, P = 0.002; rs8099917, P = 0.001), in addition to a high baseline viral load (rs12979860, P = 1.4 × 10−5; rs8099917, P = 7.3 × 10−6). Conclusion: Polymorphisms near the IL28B gene show association with rapid viral response but not sustained viral response to PEG‐IFN/ribavirin therapy in HCV genotype 3‐infected patients. (HEPATOLOGY 2011;)


Nature Biotechnology | 2015

Cholangiocytes derived from human induced pluripotent stem cells for disease modeling and drug validation

Fotios Sampaziotis; Miguel Cardoso de Brito; Pedro Madrigal; Alessandro Bertero; Kourosh Saeb-Parsy; Filipa Soares; Elisabeth Schrumpf; Espen Melum; Tom H. Karlsen; J. Andrew Bradley; W. Gelson; Susan E. Davies; Alastair Baker; Arthur Kaser; Graeme J. M. Alexander; Nicholas Hannan; Ludovic Vallier

The study of biliary disease has been constrained by a lack of primary human cholangiocytes. Here we present an efficient, serum-free protocol for directed differentiation of human induced pluripotent stem cells into cholangiocyte-like cells (CLCs). CLCs show functional characteristics of cholangiocytes, including bile acids transfer, alkaline phosphatase activity, γ-glutamyl-transpeptidase activity and physiological responses to secretin, somatostatin and vascular endothelial growth factor. We use CLCs to model in vitro key features of Alagille syndrome, polycystic liver disease and cystic fibrosis (CF)-associated cholangiopathy. Furthermore, we use CLCs generated from healthy individuals and patients with polycystic liver disease to reproduce the effects of the drugs verapamil and octreotide, and we show that the experimental CF drug VX809 rescues the disease phenotype of CF cholangiopathy in vitro. Our differentiation protocol will facilitate the study of biological mechanisms controlling biliary development, as well as disease modeling and drug screening.


Hepatology | 2011

Three ulcerative colitis susceptibility loci are associated with primary sclerosing cholangitis and indicate a role for IL2, REL and CARD9

Marcel Janse; Laetitia E. Lamberts; Lude Franke; Soumya Raychaudhuri; Eva Ellinghaus; Kirsten Muri Boberg; Espen Melum; Trine Folseraas; Erik Schrumpf; Annika Bergquist; Einar Björnsson; Jingyuan Fu; Harm-Jan Westra; Harry J.M. Groen; Rudolf S. N. Fehrmann; Joanna Smolonska; Leonard H. van den Berg; Roel A. Ophoff; Robert J. Porte; Tobias J. Weismüller; Jochen Wedemeyer; Christoph Schramm; Martina Sterneck; Rainer Günther; Felix Braun; Severine Vermeire; Liesbet Henckaerts; Cisca Wijmenga; Cyriel Y. Ponsioen; Stefan Schreiber

Primary sclerosing cholangitis (PSC) is a chronic cholestatic liver disease characterized by inflammation and fibrosis of the bile ducts. Both environmental and genetic factors contribute to its pathogenesis. To further clarify its genetic background, we investigated susceptibility loci recently identified for ulcerative colitis (UC) in a large cohort of 1,186 PSC patients and 1,748 controls. Single nucleotide polymorphisms (SNPs) tagging 13 UC susceptibility loci were initially genotyped in 854 PSC patients and 1,491 controls from Benelux (331 cases, 735 controls), Germany (265 cases, 368 controls), and Scandinavia (258 cases, 388 controls). Subsequently, a joint analysis was performed with an independent second Scandinavian cohort (332 cases, 257 controls). SNPs at chromosomes 2p16 (P‐value 4.12 × 10−4), 4q27 (P‐value 4.10 × 10−5), and 9q34 (P‐value 8.41 × 10−4) were associated with PSC in the joint analysis after correcting for multiple testing. In PSC patients without inflammatory bowel disease (IBD), SNPs at 4q27 and 9q34 were nominally associated (P < 0.05). We applied additional in silico analyses to identify likely candidate genes at PSC susceptibility loci. To identify nonrandom, evidence‐based links we used GRAIL (Gene Relationships Across Implicated Loci) analysis showing interconnectivity between genes in six out of in total nine PSC‐associated regions. Expression quantitative trait analysis from 1,469 Dutch and UK individuals demonstrated that five out of nine SNPs had an effect on cis‐gene expression. These analyses prioritized IL2, CARD9, and REL as novel candidates. Conclusion: We have identified three UC susceptibility loci to be associated with PSC, harboring the putative candidate genes REL, IL2, and CARD9. These results add to the scarce knowledge on the genetic background of PSC and imply an important role for both innate and adaptive immunological factors. (HEPATOLOGY 2011;)


Hepatology | 2013

Genome-Wide Association Analysis in Primary Sclerosing Cholangitis and Ulcerative Colitis Identifies Risk Loci at GPR35 and TCF4

David Ellinghaus; Trine Folseraas; Kristian Holm; Eva Ellinghaus; Espen Melum; Tobias Balschun; Jon K. Laerdahl; Alexey Shiryaev; Daniel Gotthardt; Tobias J. Weismüller; Christoph Schramm; Michael Wittig; Annika Bergquist; Einar Björnsson; Hanns-Ulrich Marschall; Morten H. Vatn; Andreas Teufel; Christian Rust; Christian Gieger; H-Erich Wichmann; Heiko Runz; Martina Sterneck; Christian Rupp; Felix Braun; Rinse K. Weersma; Cisca Wijmenga; Cyriel Y. Ponsioen; Christopher G. Mathew; Paul Rutgeerts; Severine Vermeire

Approximately 60%‐80% of patients with primary sclerosing cholangitis (PSC) have concurrent ulcerative colitis (UC). Previous genome‐wide association studies (GWAS) in PSC have detected a number of susceptibility loci that also show associations in UC and other immune‐mediated diseases. We aimed to systematically compare genetic associations in PSC with genotype data in UC patients with the aim of detecting new susceptibility loci for PSC. We performed combined analyses of GWAS for PSC and UC comprising 392 PSC cases, 987 UC cases, and 2,977 controls and followed up top association signals in an additional 1,012 PSC cases, 4,444 UC cases, and 11,659 controls. We discovered novel genome‐wide significant associations with PSC at 2q37 [rs3749171 at G‐protein‐coupled receptor 35 (GPR35); P = 3.0 × 10−9 in the overall study population, combined odds ratio [OR] and 95% confidence interval [CI] of 1.39 (1.24‐1.55)] and at 18q21 [rs1452787 at transcription factor 4 (TCF4); P = 2.61 × 10−8, OR (95% CI) = 0.75 (0.68‐0.83)]. In addition, several suggestive PSC associations were detected. The GPR35 rs3749171 is a missense single nucleotide polymorphism resulting in a shift from threonine to methionine. Structural modeling showed that rs3749171 is located in the third transmembrane helix of GPR35 and could possibly alter efficiency of signaling through the GPR35 receptor. Conclusion: By refining the analysis of a PSC GWAS by parallel assessments in a UC GWAS, we were able to detect two novel risk loci at genome‐wide significance levels. GPR35 shows associations in both UC and PSC, whereas TCF4 represents a PSC risk locus not associated with UC. Both loci may represent previously unexplored aspects of PSC pathogenesis. (HEPATOLOGY 2013;58:1074–1083)


BMC Bioinformatics | 2010

SNPexp - A web tool for calculating and visualizing correlation between HapMap genotypes and gene expression levels

Kristian Holm; Espen Melum; Andre Franke; Tom H. Karlsen

BackgroundExpression levels for 47294 transcripts in lymphoblastoid cell lines from all 270 HapMap phase II individuals, and genotypes (both HapMap phase II and III) of 3.96 million single nucleotide polymorphisms (SNPs) in the same individuals are publicly available. We aimed to generate a user-friendly web based tool for visualization of the correlation between SNP genotypes within a specified genomic region and a gene of interest, which is also well-known as an expression quantitative trait locus (eQTL) analysis.ResultsSNPexp is implemented as a server-side script, and publicly available on this website: http://tinyurl.com/snpexp. Correlation between genotype and transcript expression levels are calculated by performing linear regression and the Wald test as implemented in PLINK and visualized using the UCSC Genome Browser. Validation of SNPexp using previously published eQTLs yielded comparable results.ConclusionsSNPexp provides a convenient and platform-independent way to calculate and visualize the correlation between HapMap genotypes within a specified genetic region anywhere in the genome and gene expression levels. This allows for investigation of both cis and trans effects. The web interface and utilization of publicly available and widely used software resources makes it an attractive supplement to more advanced bioinformatic tools. For the advanced user the program can be used on a local computer on custom datasets.


World Journal of Gastroenterology | 2012

Recurrence and rejection in liver transplantation for primary sclerosing cholangitis.

Tom H. Karlsen; Espen Melum

Primary sclerosing cholangitis (PSC) is a chronic progressive inflammatory disease affecting the bile ducts, leading to fibrosis and eventually cirrhosis in most patients. Its etiology is unknown and so far no effective medical therapy is available. Liver transplantation (LTX) is the only curative treatment and at present PSC is the main indication for LTX in the Scandinavian countries. Close to half of the PSC patients experience one or more episodes of acute cellular rejection (ACR) following transplantation and approximately 1/5 of the transplanted patients develop recurrent disease in the graft. In addition, some reports indicate that ACR early after LTX for PSC can influence the risk for recurrent disease. For these important post-transplantation entities affecting PSC patients, we have reviewed the current literature on epidemiology, pathogenesis, treatment and the possible influence of rejection on the risk of recurrent disease in the allograft.


Hepatology | 2010

The utility of genome-wide association studies in hepatology†

Tom H. Karlsen; Espen Melum; Andre Franke

Over the last 4 years, more than 450 genome‐wide association studies (GWAS) have been successfully performed in a variety of human traits, of which approximately 2% relates to the field of hepatology. Whereas the many robust susceptibility gene findings have provided insight into fundamental physiological aspects of the phenotypes that have been studied, the widespread application has also revealed important limitations of the GWAS design. This review aims to systematically summarize both the strengths and the weaknesses of GWAS, as well as underscore important experiences made in model diseases outside the field of hepatology. By reviewing the GWAS performed in hepatology so far on this broader background, extensions and guidelines for the rational application of the study design in hepatology are proposed. (Hepatology 2010)

Collaboration


Dive into the Espen Melum's collaboration.

Top Co-Authors

Avatar

Tom H. Karlsen

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Annika Bergquist

Karolinska University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kristian Holm

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Erik Schrumpf

Oslo University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge