Network


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

Hotspot


Dive into the research topics where Hanne Sørmo Sorte is active.

Publication


Featured researches published by Hanne Sørmo Sorte.


Blood | 2015

Early-onset lymphoproliferation and autoimmunity caused by germline STAT3 gain-of-function mutations.

Joshua D. Milner; Tiphanie P. Vogel; Lisa R. Forbes; Chi A. Ma; Asbjørg Stray-Pedersen; Julie E. Niemela; Jonathan J. Lyons; Karin R. Engelhardt; Yu Zhang; Nermina Topcagic; Elisha D. O. Roberson; Helen F. Matthews; James W. Verbsky; Trivikram Dasu; Alexander Vargas-Hernández; Nidhy P. Varghese; Kenneth L. McClain; Lina Karam; Karen Nahmod; George Makedonas; Emily M. Mace; Hanne Sørmo Sorte; Gøri Perminow; V. Koneti Rao; Michael P. O’Connell; Susan Price; Helen C. Su; Morgan Butrick; Joshua McElwee; Jason D. Hughes

Germline loss-of-function mutations in the transcription factor signal transducer and activator of transcription 3 (STAT3) cause immunodeficiency, whereas somatic gain-of-function mutations in STAT3 are associated with large granular lymphocytic leukemic, myelodysplastic syndrome, and aplastic anemia. Recently, germline mutations in STAT3 have also been associated with autoimmune disease. Here, we report on 13 individuals from 10 families with lymphoproliferation and early-onset solid-organ autoimmunity associated with 9 different germline heterozygous mutations in STAT3. Patients exhibited a variety of clinical features, with most having lymphadenopathy, autoimmune cytopenias, multiorgan autoimmunity (lung, gastrointestinal, hepatic, and/or endocrine dysfunction), infections, and short stature. Functional analyses demonstrate that these mutations confer a gain-of-function in STAT3 leading to secondary defects in STAT5 and STAT1 phosphorylation and the regulatory T-cell compartment. Treatment targeting a cytokine pathway that signals through STAT3 led to clinical improvement in 1 patient, suggesting a potential therapeutic option for such patients. These results suggest that there is a broad range of autoimmunity caused by germline STAT3 gain-of-function mutations, and that hematologic autoimmunity is a major component of this newly described disorder. Some patients for this study were enrolled in a trial registered at www.clinicaltrials.gov as #NCT00001350.


The New England Journal of Medicine | 2016

Loss of B Cells in Patients with Heterozygous Mutations in IKAROS.

Hye Sun Kuehn; Bertrand Boisson; Charlotte Cunningham-Rundles; Janine Reichenbach; Asbjørg Stray-Pedersen; Erwin W. Gelfand; Patrick Maffucci; Keith R. Pierce; Jordan K. Abbott; Karl V. Voelkerding; Sarah T. South; Nancy H. Augustine; Jeana S. Bush; William K. Dolen; Betty B. Wray; Yuval Itan; Aurélie Cobat; Hanne Sørmo Sorte; Sundar Ganesan; Seraina Prader; Thomas B. Martins; Monica G. Lawrence; Jordan S. Orange; Katherine R. Calvo; Julie E. Niemela; Jean-Laurent Casanova; Thomas A. Fleisher; Harry R. Hill; Attila Kumánovics; Mary Ellen Conley

BACKGROUND Common variable immunodeficiency (CVID) is characterized by late-onset hypogammaglobulinemia in the absence of predisposing factors. The genetic cause is unknown in the majority of cases, and less than 10% of patients have a family history of the disease. Most patients have normal numbers of B cells but lack plasma cells. METHODS We used whole-exome sequencing and array-based comparative genomic hybridization to evaluate a subset of patients with CVID and low B-cell numbers. Mutant proteins were analyzed for DNA binding with the use of an electrophoretic mobility-shift assay (EMSA) and confocal microscopy. Flow cytometry was used to analyze peripheral-blood lymphocytes and bone marrow aspirates. RESULTS Six different heterozygous mutations in IKZF1, the gene encoding the transcription factor IKAROS, were identified in 29 persons from six families. In two families, the mutation was a de novo event in the proband. All the mutations, four amino acid substitutions, an intragenic deletion, and a 4.7-Mb multigene deletion involved the DNA-binding domain of IKAROS. The proteins bearing missense mutations failed to bind target DNA sequences on EMSA and confocal microscopy; however, they did not inhibit the binding of wild-type IKAROS. Studies in family members showed progressive loss of B cells and serum immunoglobulins. Bone marrow aspirates in two patients had markedly decreased early B-cell precursors, but plasma cells were present. Acute lymphoblastic leukemia developed in 2 of the 29 patients. CONCLUSIONS Heterozygous mutations in the transcription factor IKAROS caused an autosomal dominant form of CVID that is associated with a striking decrease in B-cell numbers. (Funded by the National Institutes of Health and others.).


The Journal of Allergy and Clinical Immunology | 2017

Primary immunodeficiency diseases: Genomic approaches delineate heterogeneous Mendelian disorders

Asbjørg Stray-Pedersen; Hanne Sørmo Sorte; Pubudu Saneth Samarakoon; Tomasz Gambin; Ivan K. Chinn; Zeynep Coban Akdemir; Hans Christian Erichsen; Lisa R. Forbes; Shen Gu; Bo Yuan; Shalini N. Jhangiani; Donna M. Muzny; Olaug K. Rødningen; Ying Sheng; Sarah K. Nicholas; Lenora M. Noroski; Filiz O. Seeborg; Carla M. Davis; Debra L. Canter; Emily M. Mace; Timothy J. Vece; Carl E. Allen; Harshal Abhyankar; Philip M. Boone; Christine R. Beck; Wojciech Wiszniewski; Børre Fevang; Pål Aukrust; Geir E. Tjønnfjord; Tobias Gedde-Dahl

Background: Primary immunodeficiency diseases (PIDDs) are clinically and genetically heterogeneous disorders thus far associated with mutations in more than 300 genes. The clinical phenotypes derived from distinct genotypes can overlap. Genetic etiology can be a prognostic indicator of disease severity and can influence treatment decisions. Objective: We sought to investigate the ability of whole‐exome screening methods to detect disease‐causing variants in patients with PIDDs. Methods: Patients with PIDDs from 278 families from 22 countries were investigated by using whole‐exome sequencing. Computational copy number variant (CNV) prediction pipelines and an exome‐tiling chromosomal microarray were also applied to identify intragenic CNVs. Analytic approaches initially focused on 475 known or candidate PIDD genes but were nonexclusive and further tailored based on clinical data, family history, and immunophenotyping. Results: A likely molecular diagnosis was achieved in 110 (40%) unrelated probands. Clinical diagnosis was revised in about half (60/110) and management was directly altered in nearly a quarter (26/110) of families based on molecular findings. Twelve PIDD‐causing CNVs were detected, including 7 smaller than 30 Kb that would not have been detected with conventional diagnostic CNV arrays. Conclusion: This high‐throughput genomic approach enabled detection of disease‐related variants in unexpected genes; permitted detection of low‐grade constitutional, somatic, and revertant mosaicism; and provided evidence of a mutational burden in mixed PIDD immunophenotypes.


BMC Genomics | 2014

Identification of copy number variants from exome sequence data

Pubudu Saneth Samarakoon; Hanne Sørmo Sorte; Bjørn Evert Kristiansen; Tove Skodje; Ying Sheng; Geir E. Tjønnfjord; Barbro Stadheim; Asbjørg Stray-Pedersen; Olaug K. Rødningen; Robert Lyle

BackgroundWith advances in next generation sequencing technologies and genomic capture techniques, exome sequencing has become a cost-effective approach for mutation detection in genetic diseases. However, computational prediction of copy number variants (CNVs) from exome sequence data is a challenging task. Whilst numerous programs are available, they have different sensitivities, and have low sensitivity to detect smaller CNVs (1–4 exons). Additionally, exonic CNV discovery using standard aCGH has limitations due to the low probe density over exonic regions. The goal of our study was to develop a protocol to detect exonic CNVs (including shorter CNVs that cover 1–4 exons), combining computational prediction algorithms and a high-resolution custom CGH array.ResultsWe used six published CNV prediction programs (ExomeCNV, CONTRA, ExomeCopy, ExomeDepth, CoNIFER, XHMM) and an in-house modification to ExomeCopy and ExomeDepth (ExCopyDepth) for computational CNV prediction on 30 exomes from the 1000 genomes project and 9 exomes from primary immunodeficiency patients. CNV predictions were tested using a custom CGH array designed to capture all exons (exaCGH). After this validation, we next evaluated the computational prediction of shorter CNVs. ExomeCopy and the in-house modified algorithm, ExCopyDepth, showed the highest capability in detecting shorter CNVs. Finally, the performance of each computational program was assessed by calculating the sensitivity and false positive rate.ConclusionsIn this paper, we assessed the ability of 6 computational programs to predict CNVs, focussing on short (1–4 exon) CNVs. We also tested these predictions using a custom array targeting exons. Based on these results, we propose a protocol to identify and confirm shorter exonic CNVs combining computational prediction algorithms and custom aCGH experiments.


European Journal of Human Genetics | 2012

Diagnosis by sequencing: correction of misdiagnosis from FSHD2 to LGMD2A by whole-exome analysis

Andreas Leidenroth; Hanne Sørmo Sorte; Gregor D. Gilfillan; Melanie Ehrlich; Robert Lyle; Jane E. Hewitt

We studied and validated facioscapulohumeral muscular dystrophy (FSHD) samples from patients without a D4Z4 contraction (FSHD2 or ‘phenotypic FSHD’). For this, we developed non-radioactive protocols to test D4Z4 allele constitution and DNA methylation, and applied these to samples from the Coriell Institute Cell Repository. The D4Z4 sizing showed two related subjects to have classic chromosome 4 contraction-dependent FSHD1. A third sample (GM17726) did not have a short chromosome 4 fragment, and had been assigned as non-4q FSHD (FSHD2). We tested D4Z4 haplotype and methylation for this individual but found both to be inconsistent with this diagnosis. Using exome sequencing, we identified two known pathogenic mutations in CAPN3 (Arg490Gln and Thr184Argfs*36), indicating a case of LGMD2A rather than FSHD. Our study shows how a wrong diagnosis can easily be corrected by whole-exome sequencing by constraining the variant analysis to candidate genes after the data have been generated. This new way of ‘diagnosis by sequencing’ is likely to become common place in genetic diagnostic laboratories. We also publish a digoxigenin-labeled Southern protocol to test D4Z4 methylation. Our data supports hypomethylation as a good epigenetic predictor for FSHD2. The non-radioactive protocol will help to make this assay more accessible to clinical diagnostic laboratories and the wider FSHD research community.


Clinical and Experimental Immunology | 2014

Vaccine-associated varicella and rubella infections in severe combined immunodeficiency with isolated CD4 lymphocytopenia and mutations in IL7R detected by tandem whole exome sequencing and chromosomal microarray

D. K. Bayer; Caridad Martinez; Hanne Sørmo Sorte; Lisa R. Forbes; G. J. Demmler-Harrison; I. C. Hanson; N. M. Pearson; Lenora M. Noroski; S. R. Zaki; W. J. Bellini; Magalie S. Leduc; Yaping Yang; Christine M. Eng; Ankita Patel; Olaug K. Rødningen; Donna M. Muzny; Richard A. Gibbs; Ian M. Campbell; Chad A. Shaw; M. W. Baker; Victor Wei Zhang; James R. Lupski; Jordan S. Orange; Filiz O. Seeborg; Asbjørg Stray-Pedersen

In areas without newborn screening for severe combined immunodeficiency (SCID), disease‐defining infections may lead to diagnosis, and in some cases, may not be identified prior to the first year of life. We describe a female infant who presented with disseminated vaccine‐acquired varicella (VZV) and vaccine‐acquired rubella infections at 13 months of age. Immunological evaluations demonstrated neutropenia, isolated CD4 lymphocytopenia, the presence of CD8+ T cells, poor lymphocyte proliferation, hypergammaglobulinaemia and poor specific antibody production to VZV infection and routine immunizations. A combination of whole exome sequencing and custom‐designed chromosomal microarray with exon coverage of primary immunodeficiency genes detected compound heterozygous mutations (one single nucleotide variant and one intragenic copy number variant involving one exon) within the IL7R gene. Mosaicism for wild‐type allele (20–30%) was detected in pretransplant blood and buccal DNA and maternal engraftment (5–10%) demonstrated in pretransplant blood DNA. This may be responsible for the patients unusual immunological phenotype compared to classical interleukin (IL)‐7Rα deficiency. Disseminated VZV was controlled with anti‐viral and immune‐based therapy, and umbilical cord blood stem cell transplantation was successful. Retrospectively performed T cell receptor excision circle (TREC) analyses completed on neonatal Guthrie cards identified absent TREC. This case emphasizes the danger of live viral vaccination in severe combined immunodeficiency (SCID) patients and the importance of newborn screening to identify patients prior to high‐risk exposures. It also illustrates the value of aggressive pathogen identification and treatment, the influence newborn screening can have on morbidity and mortality and the significant impact of newer genomic diagnostic tools in identifying the underlying genetic aetiology for SCID patients.


Psychiatric Genetics | 2013

Copy number variation findings among 50 children and adolescents with autism spectrum disorder.

Hanne Sørmo Sorte; Elen Gjevik; Eili Sponheim; Kristin Eiklid; Olaug K. Rødningen

Objectives Autism spectrum disorders (ASDs) are a heterogeneous group of neurodevelopment disorders with a complex genetic aetiology. The aim of this study was to identify copy number variations (CNVs) with a clinical significance for ASD. Materials and methods Array-based comparative genomic hybridization was applied to detect CNVs in a clinically well-characterized population of 50 children and adolescents with ASD. Results Nine CNVs with predicted clinical significance were identified among eight individuals (detection rate 16%). Three of the CNVs are recurrently associated with ASDs (15q11.2q13.1) or have been identified in ASD populations [3p14.2 and t(8;12)(p23.1;p13.31)]. The remaining regions (15q11.2, 10q21.1, Xp22.2, 16p13.3 and 22q13.1) have not been reported previously as candidate genes for ASD. Conclusion This study identified five novel CNVs among the individuals. The causal relationship between identified CNVs and the ASD phenotype is not fully established. However, the genes involved are associated with ASD and/or other neuropsychiatric disorders, or implicated in synaptic and neuronal activity, thus suggesting clinical significance. Further identification of ASD-associated CNVs is required, together with a broad clinical characterization of affected individuals to identify genotype–phenotype correlations.


BMC Genomics | 2016

cnvScan: a CNV screening and annotation tool to improve the clinical utility of computational CNV prediction from exome sequencing data

Pubudu Saneth Samarakoon; Hanne Sørmo Sorte; Asbjørg Stray-Pedersen; Olaug K. Rødningen; Torbjørn Rognes; Robert Lyle

BackgroundWith advances in next generation sequencing technology and analysis methods, single nucleotide variants (SNVs) and indels can be detected with high sensitivity and specificity in exome sequencing data. Recent studies have demonstrated the ability to detect disease-causing copy number variants (CNVs) in exome sequencing data. However, exonic CNV prediction programs have shown high false positive CNV counts, which is the major limiting factor for the applicability of these programs in clinical studies.ResultsWe have developed a tool (cnvScan) to improve the clinical utility of computational CNV prediction in exome data. cnvScan can accept input from any CNV prediction program. cnvScan consists of two steps: CNV screening and CNV annotation. CNV screening evaluates CNV prediction using quality scores and refines this using an in-house CNV database, which greatly reduces the false positive rate. The annotation step provides functionally and clinically relevant information using multiple source datasets.We assessed the performance of cnvScan on CNV predictions from five different prediction programs using 64 exomes from Primary Immunodeficiency (PIDD) patients, and identified PIDD-causing CNVs in three individuals from two different families.ConclusionsIn summary, cnvScan reduces the time and effort required to detect disease-causing CNVs by reducing the false positive count and providing annotation. This improves the clinical utility of CNV detection in exome data.


Molecular Genetics & Genomic Medicine | 2016

A potential founder variant in CARMIL2/RLTPR in three Norwegian families with warts, molluscum contagiosum, and T‐cell dysfunction

Hanne Sørmo Sorte; Liv T. Osnes; Børre Fevang; Pål Aukrust; Hans Christian Erichsen; Paul Hoff Backe; Tore G. Abrahamsen; Ole B. Kittang; Torstein Øverland; Shalini N. Jhangiani; Donna M. Muzny; Magnus Dehli Vigeland; Pubudu Saneth Samarakoon; Tomasz Gambin; Zeynep Coban Akdemir; Richard A. Gibbs; Olaug K. Rødningen; Robert Lyle; James R. Lupski; Asbjørg Stray-Pedersen

Four patients from three Norwegian families presented with a common skin phenotype of warts, molluscum contagiosum, and dermatitis since early childhood, and various other immunological features. Warts are a common manifestation of human papilloma virus (HPV), but when they are overwhelming, disseminated and/or persistent, and presenting together with other immunological features, a primary immunodeficiency disease (PIDD) may be suspected.


Molecular Syndromology | 2016

Exome Sequencing Fails to Identify the Genetic Cause of Aicardi Syndrome

Caroline Lund; Pasquale Striano; Hanne Sørmo Sorte; Pasquale Parisi; Michele Iacomino; Ying Sheng; Magnus Dehli Vigeland; Anne Marte Øye; Rikke S. Møller; Kaja Kristine Selmer; Federico Zara

Aicardi syndrome (AS) is a well-characterized neurodevelopmental disorder with an unknown etiology. In this study, we performed whole-exome sequencing in 11 female patients with the diagnosis of AS, in order to identify the disease-causing gene. In particular, we focused on detecting variants in the X chromosome, including the analysis of variants with a low number of sequencing reads, in case of somatic mosaicism. For 2 of the patients, we also sequenced the exome of the parents to search for de novo mutations. We did not identify any genetic variants likely to be damaging. Only one single missense variant was identified by the de novo analyses of the 2 trios, and this was considered benign. The failure to identify a disease gene in this study may be due to technical limitations of our study design, including the possibility that the genetic aberration leading to AS is situated in a non-exonic region or that the mutation is somatic and not detectable by our approach. Alternatively, it is possible that AS is genetically heterogeneous and that 11 patients are not sufficient to reveal the causative genes. Future studies of AS should consider designs where also non-exonic regions are explored and apply a sequencing depth so that also low-grade somatic mosaicism can be detected.

Collaboration


Dive into the Hanne Sørmo Sorte's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Robert Lyle

Oslo University Hospital

View shared research outputs
Top Co-Authors

Avatar

Donna M. Muzny

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

James R. Lupski

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar

Lisa R. Forbes

Baylor College of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tomasz Gambin

Warsaw University of Technology

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ying Sheng

Oslo University Hospital

View shared research outputs
Researchain Logo
Decentralizing Knowledge