Digitala Vetenskapliga Arkivet

Change search
Refine search result
1 - 48 of 48
CiteExportLink to result list
Permanent link
Cite
Citation style
  • apa
  • ieee
  • modern-language-association-8th-edition
  • vancouver
  • Other style
More styles
Language
  • de-DE
  • en-GB
  • en-US
  • fi-FI
  • nn-NO
  • nn-NB
  • sv-SE
  • Other locale
More languages
Output format
  • html
  • text
  • asciidoc
  • rtf
Rows per page
  • 5
  • 10
  • 20
  • 50
  • 100
  • 250
Sort
  • Standard (Relevance)
  • Author A-Ö
  • Author Ö-A
  • Title A-Ö
  • Title Ö-A
  • Publication type A-Ö
  • Publication type Ö-A
  • Issued (Oldest first)
  • Issued (Newest first)
  • Created (Oldest first)
  • Created (Newest first)
  • Last updated (Oldest first)
  • Last updated (Newest first)
  • Disputation date (earliest first)
  • Disputation date (latest first)
  • Standard (Relevance)
  • Author A-Ö
  • Author Ö-A
  • Title A-Ö
  • Title Ö-A
  • Publication type A-Ö
  • Publication type Ö-A
  • Issued (Oldest first)
  • Issued (Newest first)
  • Created (Oldest first)
  • Created (Newest first)
  • Last updated (Oldest first)
  • Last updated (Newest first)
  • Disputation date (earliest first)
  • Disputation date (latest first)
Select
The maximal number of hits you can export is 250. When you want to export more records please use the Create feeds function.
  • 1.
    Aarestrup, F. M.
    et al.
    Tech Univ Denmark, Lyngby, Denmark..
    Auffray, C.
    EISBM, Vourles, France..
    Benhabiles, N.
    Univ Paris Saclay, CEA, French Atom Energy & Alternat Energy Commiss, Direct Rech Fondamentale, F-91191 Gif Sur Yvette, France..
    Blomberg, N.
    ELIXIR, Welcome Genome Campus, Cambridge CB10 1SD, England..
    Korbel, J. O.
    European Mol Biol Lab, Genome Biol Unit, Heidelberg, Germany..
    Oksvold, Per
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH Royal Inst Technol, Sci Life Lab, Stockholm, Sweden..
    Van Oyen, H.
    Univ Sci & Technol, Dept Comp Sci, Krakow, Poland.;Univ Sci & Technol, Akad Gornizco Hutnizca, Acad Comp Ctr Cyfronet, Krakow, Poland.;Sciensano, Juliette Wystmanstr, B-1050 Brussels, Belgium..
    Towards a European health research and innovation cloud (HRIC)2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 18Article in journal (Refereed)
    Abstract [en]

    The European Union (EU) initiative on the Digital Transformation of Health and Care (Digicare) aims to provide the conditions necessary for building a secure, flexible, and decentralized digital health infrastructure. Creating a European Health Research and Innovation Cloud (HRIC) within this environment should enable data sharing and analysis for health research across the EU, in compliance with data protection legislation while preserving the full trust of the participants. Such a HRIC should learn from and build on existing data infrastructures, integrate best practices, and focus on the concrete needs of the community in terms of technologies, governance, management, regulation, and ethics requirements. Here, we describe the vision and expected benefits of digital data sharing in health research activities and present a roadmap that fosters the opportunities while answering the challenges of implementing a HRIC. For this, we put forward five specific recommendations and action points to ensure that a European HRIC: i) is built on established standards and guidelines, providing cloud technologies through an open and decentralized infrastructure; ii) is developed and certified to the highest standards of interoperability and data security that can be trusted by all stakeholders; iii) is supported by a robust ethical and legal framework that is compliant with the EU General Data Protection Regulation (GDPR); iv) establishes a proper environment for the training of new generations of data and medical scientists; and v) stimulates research and innovation in transnational collaborations through public and private initiatives and partnerships funded by the EU through Horizon 2020 and Horizon Europe.

  • 2.
    Akan, Pelin
    et al.
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Alexeyenko, Andrey
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Costea, Paul Igor
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Hedberg, Lilia
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Werne Solnestam, Beata
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Lundin, Sverker
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Hallman, Jimmie
    Lundberg, Emma
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Uhlén, Mathias
    KTH, School of Biotechnology (BIO), Proteomics. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Lundeberg, Joakim
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Comprehensive analysis of the genome transcriptome and proteome landscapes of three tumor cell lines2012In: Genome Medicine, E-ISSN 1756-994X, Vol. 4, p. 86-Article in journal (Refereed)
    Abstract [en]

    We here present a comparative genome, transcriptome and functional network analysis of three human cancer cell lines (A431, U251MG and U2OS), and investigate their relation to protein expression. Gene copy numbers significantly influenced corresponding transcript levels; their effect on protein levels was less pronounced. We focused on genes with altered mRNA and/or protein levels to identify those active in tumor maintenance. We provide comprehensive information for the three genomes and demonstrate the advantage of integrative analysis for identifying tumor-related genes amidst numerous background mutations by relating genomic variation to expression/protein abundance data and use gene networks to reveal implicated pathways.

  • 3.
    Alfirevic, Ana
    et al.
    University of Liverpool, UK.
    Gonzalez-Galarza, Faviel
    University of Liverpool, UK.
    Bell, Catherine
    University of Liverpool, UK.
    Martinsson, Klara
    University of Liverpool, UK.
    Platt, Vivien
    University of Liverpool, UK.
    Bretland, Giovanna
    University of Liverpool, UK.
    Evely, Jane
    University of Liverpool, UK.
    Lichtenfels, Maike
    University of Liverpool, UK.
    Cederbrant, Karin
    Safety Assessment, AstraZeneca, Gartuna, Södertälje, Sweden.
    French, Neil
    University of Liverpool, UK.
    Naisbitt, Dean
    University of Liverpool, UK.
    Park, B Kevin
    University of Liverpool, UK.
    Jones, Andrew R
    University of Liverpool, UK.
    Pirmohamed, Munir
    University of Liverpool, UK.
    In silico analysis of HLA associations with drug-induced liver injury: use of a HLA-genotyped DNA archive from healthy volunteers2012In: Genome Medicine, E-ISSN 1756-994X, Vol. 4, no 6, article id 51Article in journal (Refereed)
    Abstract [en]

    BACKGROUND: Drug-induced liver injury (DILI) is one of the most common adverse reactions leading to product withdrawal post-marketing. Recently, genome-wide association studies have identified a number of human leukocyte antigen (HLA) alleles associated with DILI; however, the cellular and chemical mechanisms are not fully understood.

    METHODS: To study these mechanisms, we established an HLA-typed cell archive from 400 healthy volunteers. In addition, we utilized HLA genotype data from more than four million individuals from publicly accessible repositories such as the Allele Frequency Net Database, Major Histocompatibility Complex Database and Immune Epitope Database to study the HLA alleles associated with DILI. We utilized novel in silico strategies to examine HLA haplotype relationships among the alleles associated with DILI by using bioinformatics tools such as NetMHCpan, PyPop, GraphViz, PHYLIP and TreeView.

    RESULTS: We demonstrated that many of the alleles that have been associated with liver injury induced by structurally diverse drugs (flucloxacillin, co-amoxiclav, ximelagatran, lapatinib, lumiracoxib) reside on common HLA haplotypes, which were present in populations of diverse ethnicity.

    CONCLUSIONS: Our bioinformatic analysis indicates that there may be a connection between the different HLA alleles associated with DILI caused by therapeutically and structurally different drugs, possibly through peptide binding of one of the HLA alleles that defines the causal haplotype. Further functional work, together with next-generation sequencing techniques, will be needed to define the causal alleles associated with DILI.

    Download full text (pdf)
    fulltext
  • 4.
    Auffray, Charles
    et al.
    European Institute Syst Biol and Med, France; University of Lyon, France.
    Balling, Rudi
    University of Luxembourg, Luxembourg.
    Barroso, Ines
    Wellcome Trust Sanger Institute, England.
    Bencze, Laszlo
    Semmelweis University, Hungary.
    Benson, Mikael
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Heart and Medicine Center, Allergy Center.
    Bergeron, Jay
    Pfizer Inc, MA 02139 USA.
    Bernal-Delgado, Enrique
    IACS IIS Aragon, Spain.
    Blomberg, Niklas
    EL IXIR, England.
    Bock, Christoph
    Austrian Academic Science, Austria; Medical University of Vienna, Austria; Max Planck Institute Informat, Germany.
    Conesa, Ana
    Principe Felipe Research Centre, Spain; University of Florida, FL 32610 USA.
    Del Signore, Susanna
    Bluecompan Ltd, England.
    Delogne, Christophe
    KPMG Luxembourg, Luxembourg.
    Devilee, Peter
    Leiden University, Netherlands.
    Di Meglio, Alberto
    European Org Nucl Research CERN, Switzerland.
    Eijkemans, Marinus
    University of Utrecht, Netherlands.
    Flicek, Paul
    European Bioinformat Institute EMBL EBI, England.
    Graf, Norbert
    University of Saarland, Germany.
    Grimm, Vera
    Forschungszentrum Julich, Germany.
    Guchelaar, Henk-Jan
    Leiden University, Netherlands.
    Guo, Yi-Ke
    University of London Imperial Coll Science Technology and Med, England.
    Glynne Gut, Ivo
    BIST, Spain.
    Hanbury, Allan
    TU Wien, Austria.
    Hanif, Shahid
    Assoc British Pharmaceut Ind, England.
    Hilgers, Ralf-Dieter
    University of Klinikum Aachen, Germany.
    Honrado, Angel
    SYNAPSE Research Management Partners, Spain.
    Rod Hose, D.
    University of Sheffield, England.
    Houwing-Duistermaat, Jeanine
    University of Leeds, England.
    Hubbard, Tim
    Kings Coll London, England; Genom England, England.
    Helen Janacek, Sophie
    European Bioinformat Institute EMBL EBI, England.
    Karanikas, Haralampos
    University of Athens, Greece.
    Kievits, Tim
    Vitr Healthcare Holding BV, Netherlands.
    Kohler, Manfred
    Fraunhofer Institute Molecular Biol and Appl Ecol ScreeningPor, Germany.
    Kremer, Andreas
    ITTM SA, Luxembourg.
    Lanfear, Jerry
    Pfizer Ltd, England.
    Lengauer, Thomas
    Max Planck Institute for Informatics, Saarbrucken, Germany.
    Maes, Edith
    Health Econ and Outcomes Research, Belgium.
    Meert, Theo
    Janssen Pharmaceut NV, Belgium.
    Mueller, Werner
    University of Manchester, England.
    Nickel, Dorthe
    Institute Curie, France.
    Oledzki, Peter
    Linguamat Ltd, England.
    Pedersen, Bertrand
    PwC Luxembourg, Luxembourg.
    Petkovic, Milan
    Philips, Netherlands.
    Pliakos, Konstantinos
    KU Leuven Kulak, Belgium.
    Rattray, Magnus
    University of Manchester, England.
    Redon i Mas, Josep
    University of Valencia, Spain.
    Schneider, Reinhard
    University of Luxembourg, Luxembourg.
    Sengstag, Thierry
    SIB, Switzerland; University of Basel, Switzerland.
    Serra-Picamal, Xavier
    Agency Health Qual and Assessment Catalonia AQuAS, Spain.
    Spek, Wouter
    EuroBioForum Fdn, Netherlands.
    Vaas, Lea A. I.
    Fraunhofer Institute Molecular Biol and Appl Ecol ScreeningPor, Germany.
    van Batenburg, Okker
    EuroBioForum Fdn, Netherlands.
    Vandelaer, Marc
    Integrated BioBank Luxembourg, Luxembourg.
    Varnai, Peter
    Technopolis Grp, England.
    Villoslada, Pablo
    Hospital Clin Barcelona, Spain.
    Antonio Vizcaino, Juan
    European Bioinformat Institute EMBL EBI, England.
    Peter Mary Wubbe, John
    European Platform Patients Org Science and Ind Epposi, Belgium.
    Zanetti, Gianluigi
    CRS4, Italy; BBMRI ERIC, Austria.
    Making sense of big data in health research: Towards an EU action plan2016In: Genome Medicine, E-ISSN 1756-994X, Vol. 8, no 71Article in journal (Refereed)
    Abstract [en]

    Medicine and healthcare are undergoing profound changes. Whole-genome sequencing and high-resolution imaging technologies are key drivers of this rapid and crucial transformation. Technological innovation combined with automation and miniaturization has triggered an explosion in data production that will soon reach exabyte proportions. How are we going to deal with this exponential increase in data production? The potential of "big data" for improving health is enormous but, at the same time, we face a wide range of challenges to overcome urgently. Europe is very proud of its cultural diversity; however, exploitation of the data made available through advances in genomic medicine, imaging, and a wide range of mobile health applications or connected devices is hampered by numerous historical, technical, legal, and political barriers. European health systems and databases are diverse and fragmented. There is a lack of harmonization of data formats, processing, analysis, and data transfer, which leads to incompatibilities and lost opportunities. Legal frameworks for data sharing are evolving. Clinicians, researchers, and citizens need improved methods, tools, and training to generate, analyze, and query data effectively. Addressing these barriers will contribute to creating the European Single Market for health, which will improve health arid healthcare for all Europearis.

    Download full text (pdf)
    fulltext
  • 5.
    Auffray, Charles
    et al.
    European Institute Syst Biol and Med, France; University of Lyon, France.
    Balling, Rudi
    University of Luxembourg, Luxembourg.
    Barroso, Ines
    Wellcome Trust Sanger Institute, England.
    Bencze, Laszlo
    Semmelweis University, Hungary.
    Benson, Mikael
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Heart and Medicine Center, Allergy Center.
    Bergeron, Jay
    Pfizer Inc, MA 02139 USA.
    Bernal-Delgado, Enrique
    IACS IIS Aragon, Spain.
    Blomberg, Niklas
    ELIXIR, England.
    Bock, Christoph
    Austrian Academic Science, Austria; Austrian Academic Science, Austria; Max Planck Institute Informat, Germany.
    Conesa, Ana
    Principe Felipe Research Centre, Spain; University of Florida, FL 32610 USA.
    Del Signore, Susanna
    Bluecompan Ltd, England.
    Delogne, Christophe
    KPMG Luxembourg, Luxembourg.
    Devilee, Peter
    Leiden University, Netherlands.
    Di Meglio, Alberto
    European Org Nucl Research, Switzerland.
    Eijkemans, Marinus
    University of Medical Centre Utrecht, Netherlands.
    Flicek, Paul
    EMBL EBI, England.
    Graf, Norbert
    University of Saarland, Germany.
    Grimm, Vera
    Forschungszentrum Julich, Germany.
    Guchelaar, Henk-Jan
    Leiden University, Netherlands.
    Guo, Yi-Ke
    Imperial Coll London, England.
    Glynne Gut, Ivo
    BIST, Spain.
    Hanbury, Allan
    TU Wien, Austria.
    Hanif, Shahid
    Assoc British Pharmaceut Ind, England.
    Hilgers, Ralf-Dieter
    Rhein Westfal TH Aachen, Germany.
    Honrado, Angel
    SYNAPSE Research Management Partners, Spain.
    Rod Hose, D.
    University of Sheffield, England.
    Houwing-Duistermaat, Jeanine
    University of Leeds, England.
    Hubbard, Tim
    Kings Coll London, England; Genom England, England.
    Helen Janacek, Sophie
    EMBL EBI, England.
    Karanikas, Haralampos
    University of Athens, Greece.
    Kievits, Tim
    Vitromics Healthcare Holding BV, Netherlands.
    Kohler, Manfred
    Fraunhofer Institute Molecular Biol and Appl Ecol ScreeningPor, Germany.
    Kremer, Andreas
    ITTM SA, Luxembourg.
    Lanfear, Jerry
    Pfizer Ltd, England.
    Lengauer, Thomas
    Max Planck Institute Informat, Germany.
    Maes, Edith
    Deloitte Belgium, Belgium.
    Meert, Theo
    Janssen Pharmaceut NV, Belgium.
    Muller, Werner
    University of Manchester, England.
    Nickel, Dothe
    Institute Curie, France.
    Oledzki, Peter
    Linguamat Ltd, England.
    Pedersen, Bertrand
    PwC Luxembourg, Luxembourg.
    Petkovic, Milan
    Philips, Netherlands.
    Pliakos, Konstantinos
    KU Leuven Kulak, Belgium.
    Rattray, Magnus
    University of Manchester, England.
    Redon i Mas, Josep
    University of Valencia, Spain.
    Schneider, Reinhard
    University of Luxembourg, Luxembourg.
    Sengstag, Thierry
    SIB, Switzerland; University of Basel, Switzerland.
    Serra-Picamal, Xavier
    Agency Health Qual and Assessment Catalonia AQuAS, Spain.
    Spek, Wouter
    EuroBioForum Fdn, Netherlands.
    Vaas, Lea A. I.
    Fraunhofer Institute Molecular Biol and Appl Ecol ScreeningPor, Germany.
    van Batenburg, Okker
    EuroBioForum Fdn, Netherlands.
    Vandelaer, Marc
    Integrated BioBank Luxembourg, Luxembourg.
    Varnai, Peter
    Technopolis Grp, England.
    Villoslada, Pablo
    Hospital Clin Barcelona, Spain.
    Antonio Vizcaino, Juan
    EMBL EBI, England.
    Peter Mary Wubbe, John
    Epposi, Belgium.
    Zanetti, Gianluigi
    CRS4, Italy; BBMRI ERIC, Austria.
    Correction: Making sense of big data in health research: towards an EU action plan (vol 8, pg 71, 2016)2016In: Genome Medicine, E-ISSN 1756-994X, Vol. 8, article id 118Article in journal (Other academic)
    Abstract [en]

    n/a

    Download full text (pdf)
    fulltext
  • 6.
    Bedarf, J. R.
    et al.
    Univ Bonn, Dept Neurol, Bonn, Germany.;German Ctr Neurodegenerat Dis Res DZNE, Bonn, Germany..
    Hildebrand, F.
    EMBL, Heidelberg, Germany..
    Coelho, L. P.
    EMBL, Heidelberg, Germany..
    Sunagawa, S.
    EMBL, Heidelberg, Germany.;Swiss Fed Inst Technol, Inst Microbiol, Vladimir Prelog 1-5-10, CH-8093 Zurich, Switzerland..
    Bahram, Mohammad
    Uppsala University, Disciplinary Domain of Science and Technology, Biology, Department of Organismal Biology, Systematic Biology. Univ Tartu, Inst Ecol & Earth, 40 Lai St, EE-51005 Tartu, Estonia..
    Goeser, F.
    Univ Bonn, Dept Internal Med 1, Bonn, Germany.;German Ctr Infect Res DZIF, Bonn, Germany..
    Bork, P.
    EMBL, Heidelberg, Germany.;Heidelberg Univ, MMPU, Heidelberg, Germany.;European Mol Biol Lab, Heidelberg, Germany.;Max Delbruck Ctr Mol Med, D-13125 Berlin, Germany.;Univ Wurzburg, Dept Bioinformat, D-97074 Wurzburg, Germany.;Meyerhofstr 1, D-69117 Heidelberg, Germany..
    Wüllner, U.
    Univ Bonn, Dept Neurol, Bonn, Germany.;German Ctr Neurodegenerat Dis Res DZNE, Bonn, Germany.;Sigmund Freud Str 25, D-53127 Bonn, Germany..
    Functional implications of microbial and viral gut metagenome changes in early stage L-DOPA-naive Parkinson's disease patients2017In: Genome Medicine, E-ISSN 1756-994X, Vol. 9, article id 39Article in journal (Refereed)
    Abstract [en]

    Background: Parkinson's disease (PD) presently is conceptualized as a protein aggregation disease in which pathology involves both the enteric and the central nervous system, possibly spreading from one to another via the vagus nerves. As gastrointestinal dysfunction often precedes or parallels motor symptoms, the enteric system with its vast diversity of microorganisms may be involved in PD pathogenesis. Alterations in the enteric microbial taxonomic level of L-DOPA-naive PD patients might also serve as a biomarker.

    Methods: We performed metagenomic shotgun analyses and compared the fecal microbiomes of 31 early stage, L-DOPA-naive PD patients to 28 age-matched controls.

    Results: We found increased Verrucomicrobiaceae (Akkermansia muciniphila) and unclassified Firmicutes, whereas Prevotellaceae (Prevotella copri) and Erysipelotrichaceae (Eubacterium biforme) were markedly lowered in PD samples. The observed differences could reliably separate PD from control with a ROC-AUC of 0.84. Functional analyses of the metagenomes revealed differences in microbiota metabolism in PD involving the beta-glucuronate and tryptophan metabolism. While the abundances of prophages and plasmids did not differ between PD and controls, total virus abundance was decreased in PD participants. Based on our analyses, the intake of either a MAO inhibitor, amantadine, or a dopamine agonist (which in summary relates to 90% of PD patients) had no overall influence on taxa abundance or microbial functions.

    Conclusions: Our data revealed differences of colonic microbiota and of microbiota metabolism between PD patients and controls at an unprecedented detail not achievable through 16S sequencing. The findings point to a yet unappreciated aspect of PD, possibly involving the intestinal barrier function and immune function in PD patients. The influence of the parkinsonian medication should be further investigated in the future in larger cohorts.

    Download full text (pdf)
    fulltext
  • 7.
    Björnsson, Bergthor
    et al.
    Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Surgery, Orthopaedics and Cancer Treatment, Department of Surgery in Linköping. Linköping University, Department of Biomedical and Clinical Sciences, Division of Surgery, Orthopedics and Oncology.
    Borrebaeck, Carl
    Lund Univ, Sweden.
    Elander, Nils
    Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Surgery, Orthopaedics and Cancer Treatment, Department of Oncology. Linköping University, Department of Biomedical and Clinical Sciences, Division of Surgery, Orthopedics and Oncology.
    Gasslander, Thomas
    Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Surgery, Orthopaedics and Cancer Treatment, Department of Surgery in Linköping. Linköping University, Department of Biomedical and Clinical Sciences, Division of Surgery, Orthopedics and Oncology.
    Gawel, Danuta
    Linköping University, Faculty of Medicine and Health Sciences. Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health.
    Gustafsson, Mika
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Jornsten, Rebecka
    Univ Gothenburg, Sweden; Chalmers Univ Technol, Sweden.
    Jung Lee, Eun Jung
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences. Yonsei Univ, South Korea.
    Li, Xinxiu
    Linköping University, Faculty of Medicine and Health Sciences. Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health.
    Lilja, Sandra
    Linköping University, Faculty of Medicine and Health Sciences. Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health.
    Martinez, David
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Matussek, Andreas
    Karolinska Univ Hosp, Sweden; Dept Lab Med, Sweden.
    Sandström, Per
    Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Surgery, Orthopaedics and Cancer Treatment, Department of Surgery in Linköping. Linköping University, Department of Biomedical and Clinical Sciences, Division of Surgery, Orthopedics and Oncology.
    Schäfer, Samuel
    Linköping University, Faculty of Medicine and Health Sciences. Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health.
    Stenmarker, Margaretha
    Futurum Acad Hlth and Care, Sweden; Inst Clin Sci, Sweden.
    Sun, Xiao-Feng
    Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Surgery, Orthopaedics and Cancer Treatment, Department of Oncology. Linköping University, Department of Biomedical and Clinical Sciences, Division of Surgery, Orthopedics and Oncology.
    Sysoev, Oleg
    Linköping University, Department of Computer and Information Science, The Division of Statistics and Machine Learning. Linköping University, Faculty of Arts and Sciences.
    Zhang, Huan
    Linköping University, Faculty of Medicine and Health Sciences. Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health.
    Benson, Mikael
    Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center of Paediatrics and Gynaecology and Obstetrics, H.K.H. Kronprinsessan Victorias barn- och ungdomssjukhus. Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health.
    Digital twins to personalize medicine2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 4Article in journal (Other academic)
    Abstract [en]

    Personalized medicine requires the integration and processing of vast amounts of data. Here, we propose a solution to this challenge that is based on constructing Digital Twins. These are high-resolution models of individual patients that are computationally treated with thousands of drugs to find the drug that is optimal for the patient.

    Download full text (pdf)
    fulltext
  • 8.
    Bruhn-Olszewska, Bozena
    et al.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Cancer precision medicine. Uppsala University, Science for Life Laboratory, SciLifeLab.
    Davies, Hanna
    Uppsala University, Science for Life Laboratory, SciLifeLab. Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Cancer precision medicine.
    Sarkisyan, Daniil
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Cancer precision medicine. Uppsala University, Science for Life Laboratory, SciLifeLab.
    Juhas, Ulana
    Rychlicka-Buniowska, Edyta
    Wójcik, Magdalena
    Horbacz, Monika
    Jąkalski, Marcin
    Olszewski, Paweł
    Westholm, Jakub O
    Smialowska, Agata
    Wierzba, Karol
    Torinsson Naluai, Åsa
    Jern, Niklas
    Andersson, Lars-Magnus
    Järhult, Josef D.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Sciences.
    Filipowicz, Natalia
    Tiensuu Janson, Eva
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Sciences, Endocrine Oncology.
    Rubertsson, Sten
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Surgical Sciences, Anaesthesiology and Intensive Care.
    Lipcsey, Miklós
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Surgical Sciences, Anaesthesiology and Intensive Care, Hedenstierna laboratory.
    Gisslén, Magnus
    Hultström, Michael
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Cell Biology, Integrative Physiology. Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Surgical Sciences, Anaesthesiology and Intensive Care.
    Frithiof, Robert
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Surgical Sciences, Anaesthesiology and Intensive Care.
    Dumanski, Jan P.
    Uppsala University, Science for Life Laboratory, SciLifeLab. Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology. 3P-Medicine Laboratory, Medical University of Gdańsk, Dębinki 7, 80-211, Gdańsk, Poland.
    Loss of Y in leukocytes as a risk factor for critical COVID-19 in men2022In: Genome Medicine, E-ISSN 1756-994X, Vol. 14, no 1, article id 139Article in journal (Refereed)
    Abstract [en]

    BACKGROUND: The COVID-19 pandemic, which has a prominent social and economic impact worldwide, shows a largely unexplained male bias for the severity and mortality of the disease. Loss of chromosome Y (LOY) is a risk factor candidate in COVID-19 due to its prior association with many chronic age-related diseases, and its impact on immune gene transcription.

    METHODS: Publicly available scRNA-seq data of PBMC samples derived from male patients critically ill with COVID-19 were reanalyzed, and LOY status was added to the annotated cells. We further studied LOY in whole blood for 211 COVID-19 patients treated at intensive care units (ICU) from the first and second waves of the pandemic. Of these, 139 patients were subject to cell sorting for LOY analysis in granulocytes, low-density neutrophils (LDNs), monocytes, and PBMCs.

    RESULTS: Reanalysis of available scRNA-seq data revealed LDNs and monocytes as the cell types most affected by LOY. Subsequently, DNA analysis indicated that 46%, 32%, and 29% of critically ill patients showed LOY above 5% cut-off in LDNs, granulocytes, and monocytes, respectively. Hence, the myeloid lineage that is crucial for the development of severe COVID-19 phenotype is affected by LOY. Moreover, LOY correlated with increasing WHO score (median difference 1.59%, 95% HDI 0.46% to 2.71%, p=0.025), death during ICU treatment (median difference 1.46%, 95% HDI 0.47% to 2.43%, p=0.0036), and history of vessel disease (median difference 2.16%, 95% HDI 0.74% to 3.7%, p=0.004), among other variables. In 16 recovered patients, sampled during ICU stay and 93-143 days later, LOY decreased significantly in whole blood and PBMCs. Furthermore, the number of LDNs at the recovery stage decreased dramatically (median difference 76.4 per 10,000 cell sorting events, 95% HDI 55.5 to 104, p=6e-11).

    CONCLUSIONS: We present a link between LOY and an acute, life-threatening infectious disease. Furthermore, this study highlights LOY as the most prominent clonal mutation affecting the myeloid cell lineage during emergency myelopoiesis. The correlation between LOY level and COVID-19 severity might suggest that this mutation affects the functions of monocytes and neutrophils, which could have consequences for male innate immunity.

    Download full text (pdf)
    fulltext
  • 9. Bruhn-Olszewska, Bożena
    et al.
    Davies, Hanna
    Sarkisyan, Daniil
    Juhas, Ulana
    Rychlicka-Buniowska, Edyta
    Wójcik, Magdalena
    Horbacz, Monika
    Jąkalski, Marcin
    Olszewski, Paweł
    Orzechowski Westholm, Jakub
    Stockholm University, Faculty of Science, Department of Biochemistry and Biophysics. Stockholm University, Science for Life Laboratory (SciLifeLab).
    Smialowska, Agata
    Stockholm University, Faculty of Science, Department of Biochemistry and Biophysics. Stockholm University, Science for Life Laboratory (SciLifeLab).
    Wierzba, Karol
    Naluai, Åsa Torinsson
    Jern, Niklas
    Andersson, Lars-Magnus
    Järhult, Josef D.
    Filipowicz, Natalia
    Janson, Eva Tiensuu
    Rubertsson, Sten
    Lipcsey, Miklós
    Gisslén, Magnus
    Hultström, Michael
    Frithiof, Robert
    Dumanski, Jan P.
    Loss of Y in leukocytes as a risk factor for critical COVID-19 in men2022In: Genome Medicine, E-ISSN 1756-994X, Vol. 14, no 1, article id 139Article in journal (Refereed)
    Abstract [en]

    Background: The COVID-19 pandemic, which has a prominent social and economic impact worldwide, shows a largely unexplained male bias for the severity and mortality of the disease. Loss of chromosome Y (LOY) is a risk factor candidate in COVID-19 due to its prior association with many chronic age-related diseases, and its impact on immune gene transcription.

    Methods: Publicly available scRNA-seq data of PBMC samples derived from male patients critically ill with COVID-19 were reanalyzed, and LOY status was added to the annotated cells. We further studied LOY in whole blood for 211 COVID-19 patients treated at intensive care units (ICU) from the first and second waves of the pandemic. Of these, 139 patients were subject to cell sorting for LOY analysis in granulocytes, low-density neutrophils (LDNs), monocytes, and PBMCs.

    Results: Reanalysis of available scRNA-seq data revealed LDNs and monocytes as the cell types most affected by LOY. Subsequently, DNA analysis indicated that 46%, 32%, and 29% of critically ill patients showed LOY above 5% cut-off in LDNs, granulocytes, and monocytes, respectively. Hence, the myeloid lineage that is crucial for the development of severe COVID-19 phenotype is affected by LOY. Moreover, LOY correlated with increasing WHO score (median difference 1.59%, 95% HDI 0.46% to 2.71%, p=0.025), death during ICU treatment (median difference 1.46%, 95% HDI 0.47% to 2.43%, p=0.0036), and history of vessel disease (median difference 2.16%, 95% HDI 0.74% to 3.7%, p=0.004), among other variables. In 16 recovered patients, sampled during ICU stay and 93–143 days later, LOY decreased significantly in whole blood and PBMCs. Furthermore, the number of LDNs at the recovery stage decreased dramatically (median difference 76.4 per 10,000 cell sorting events, 95% HDI 55.5 to 104, p=6e−11).

    Conclusions: We present a link between LOY and an acute, life-threatening infectious disease. Furthermore, this study highlights LOY as the most prominent clonal mutation affecting the myeloid cell lineage during emergency myelopoiesis. The correlation between LOY level and COVID-19 severity might suggest that this mutation affects the functions of monocytes and neutrophils, which could have consequences for male innate immunity.

  • 10.
    Clermont, Gilles
    et al.
    University of Pittsburgh School of Medicine, PA , USA.
    Auffray, Charles
    CNRS Institute of Biological Sciences, Villejuif Cedex, France.
    Moreau, Yves
    K.U. Leuven, ESAT/SCD, Leuven-Heverlee, Belgium.
    Rocke, David M
    University of California, Davis, USA.
    Dalevi, Daniel
    Chalmers and Göteborg University, Sweden.
    Dubhashi, Devdatt
    Chalmers and Göteborg University, Sweden.
    Marshall, Dana R
    Meharry Medical College, Nashville, TN , USA.
    Raasch, Peter
    University of Rostock, Germany.
    Dehne, Frank
    Carleton University, Ottawa, Ontario, Canada.
    Provero, Paolo
    University of Torino, Italy .
    Tegner, Jesper
    Karolinska Universitetssjukhuset, Solna, Stockholm, Sweden.
    Aronow, Bruce J
    University of Cincinnati, OH, USA.
    Langston, Michael A
    University of Tennessee, Knoxville, USA.
    Benson, Mikael
    The Unit for Clinical Systems Biology, The Queen Silvia Children's Hospital, Gothenburg, Sweden.
    Bridging the gap between systems biology and medicine2009In: Genome Medicine, E-ISSN 1756-994X, Vol. 1, no 9Article in journal (Refereed)
    Abstract [en]

    Systems biology has matured considerably as a discipline over the last decade, yet some of the key challenges separating current research efforts in systems biology and clinically useful results are only now becoming apparent. As these gaps are better defined, the new discipline of systems medicine is emerging as a translational extension of systems biology. How is systems medicine defined? What are relevant ontologies for systems medicine? What are the key theoretic and methodologic challenges facing computational disease modeling? How are inaccurate and incomplete data, and uncertain biologic knowledge best synthesized in useful computational models? Does network analysis provide clinically useful insight? We discuss the outstanding difficulties in translating a rapidly growing body of data into knowledge usable at the bedside. Although core-specific challenges are best met by specialized groups, it appears fundamental that such efforts should be guided by a roadmap for systems medicine drafted by a coalition of scientists from the clinical, experimental, computational, and theoretic domains.

    Download full text (pdf)
    fulltext
  • 11.
    Denkert, Carsten
    et al.
    Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
    Bucher, Elmar
    Biotechnology for Health and Well-being, VTT Technical Research Centre of Finland, Espoo and Turku, Finland.
    Hilvo, Mika
    Biotechnology for Health and Well-being, VTT Technical Research Centre of Finland, Espoo and Turku, Finland.
    Salek, Reza
    Department of Biochemistry, University of Cambridge, Cambridge, UK.
    Oresic, Matej
    Biotechnology for Health and Well-being, VTT Technical Research Centre of Finland, Espoo and Turku, Finland.
    Griffin, Julian
    Department of Biochemistry, University of Cambridge, Cambridge, UK.
    Brockmöller, Scarlet
    Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
    Klauschen, Frederick
    Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
    Loibl, Sibylle
    German Breast Group, GBG-Forschungs GmbH, Neu-Isenburg, Germany.
    Barupal, Dinesh Kumar
    Genome Center, University of California, Davis CA, USA.
    Budczies, Jan
    Institute of Pathology, Charité - Universitätsmedizin Berlin, Berlin, Germany.
    Iljin, Kristiina
    Biotechnology for Health and Well-being, VTT Technical Research Centre of Finland, Espoo and Turku, Finland.
    Nekljudova, Valentina
    German Breast Group, GBG-Forschungs GmbH, Neu-Isenburg, Germany.
    Fiehn, Oliver
    Genome Center, University of California, Davis CA, USA.
    Metabolomics of human breast cancer: new approaches for tumor typing and biomarker discovery2012In: Genome Medicine, E-ISSN 1756-994X, Vol. 4, no 4, article id 37Article, review/survey (Refereed)
    Abstract [en]

    Breast cancer is the most common cancer in women worldwide, and the development of new technologies for better understanding of the molecular changes involved in breast cancer progression is essential. Metabolic changes precede overt phenotypic changes, because cellular regulation ultimately affects the use of small-molecule substrates for cell division, growth or environmental changes such as hypoxia. Differences in metabolism between normal cells and cancer cells have been identified. Because small alterations in enzyme concentrations or activities can cause large changes in overall metabolite levels, the metabolome can be regarded as the amplified output of a biological system. The metabolome coverage in human breast cancer tissues can be maximized by combining different technologies for metabolic profiling. Researchers are investigating alterations in the steady state concentrations of metabolites that reflect amplified changes in genetic control of metabolism. Metabolomic results can be used to classify breast cancer on the basis of tumor biology, to identify new prognostic and predictive markers and to discover new targets for future therapeutic interventions. Here, we examine recent results, including those from the European FP7 project METAcancer consortium, that show that integrated metabolomic analyses can provide information on the stage, subtype and grade of breast tumors and give mechanistic insights. We predict an intensified use of metabolomic screens in clinical and preclinical studies focusing on the onset and progression of tumor development.

  • 12.
    Feuk, Lars
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Genetics and Pathology.
    Inversion variants in the human genome: role in disease and genome architecture2010In: Genome Medicine, E-ISSN 1756-994X, Vol. 2, no 2, article id 11Article in journal (Refereed)
    Abstract [en]

    Significant advances have been made over the past 5 years in mapping and characterizing structural variation in the human genome. Despite this progress, our understanding of inversion variants is still very restricted. While unbalanced variants such as copy number variations can be mapped using array-based approaches, strategies for characterization of inversion variants have been limited and underdeveloped. Traditional cytogenetic approaches have long been able to identify microscopic inversion events, but discovery of submicroscopic events has remained elusive and largely ignored. With the advent of paired-end sequencing approaches, it is now possible to map inversions across the human genome. Based on the paired-end sequencing studies published to date, it is now feasible to make a first map of inversions across the human genome and to use this map to explore the characteristics and distribution of this form of variation. The current map of inversions indicates that many remain to be identified, especially in the smaller size ranges. This review provides an overview of the current knowledge about human inversions and their contribution to human phenotypes. Further characterization of inversions should be considered as an important step towards a deeper understanding of human variation and genome dynamics.

    Download full text (pdf)
    fulltext
  • 13.
    Gawel, Danuta
    et al.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Serra I Musach, Jordi
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Lilja, Sandra
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Aagesen, Jesper
    Reg Jonkoping Cty, Sweden.
    Arenas, Alex
    Univ Rovira and Virgili, Spain.
    Asking, Bengt
    Reg Jonkoping Cty, Sweden.
    Bengner, Malin
    Reg Jonkoping Cty, Sweden.
    Bjorkander, Janne
    Reg Jonkoping Cty, Sweden.
    Biggs, Sophie
    Linköping University, Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Science. Linköping University, Faculty of Medicine and Health Sciences.
    Ernerudh, Jan
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Diagnostics, Department of Clinical Immunology and Transfusion Medicine.
    Hjortswang, Henrik
    Linköping University, Department of Health, Medicine and Caring Sciences, Division of Diagnostics and Specialist Medicine. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Surgery, Orthopaedics and Cancer Treatment, Mag- tarmmedicinska kliniken.
    Karlsson, Jan-Erik
    Linköping University, Department of Health, Medicine and Caring Sciences, Division of Diagnostics and Specialist Medicine. Linköping University, Faculty of Medicine and Health Sciences. Reg Jonkoping Cty, Sweden.
    Köpsen, Mattias
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Jung Lee, Eun Jung
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences. Yonsei Univ, South Korea.
    Lentini, Antonio
    Linköping University, Department of Physics, Chemistry and Biology, Biology. Linköping University, Faculty of Science & Engineering.
    Li, Xinxiu
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Magnusson, Mattias
    Linköping University, Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Science. Linköping University, Faculty of Medicine and Health Sciences.
    Martinez, David
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Matussek, Andreas
    Reg Jonkoping Cty, Sweden; Karolinska Inst, Sweden; Karolinska Univ Hosp, Sweden.
    Nestor, Colm
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Schäfer, Samuel
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Seifert, Oliver
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Cell Biology. Linköping University, Faculty of Medicine and Health Sciences. Reg Jonkoping Cty, Sweden.
    Sonmez, Ceylan
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Molecular Medicine and Virology. Linköping University, Faculty of Medicine and Health Sciences.
    Stjernman, Henrik
    Reg Jonkoping Cty, Sweden.
    Tjärnberg, Andreas
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Wu, Simon
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Åkesson, Karin
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences. Reg Jonkoping Cty, Sweden.
    Shalek, Alex K.
    MIT, MA 02139 USA; MIT, MA 02139 USA; MIT, MA 02139 USA; Broad Inst MIT and Harvard, MA 02142 USA; Ragon Inst MGH MIT and Harvard, MA USA.
    Stenmarker, Margaretha
    Reg Jonkoping Cty, Sweden; Inst Clin Sci, Sweden.
    Zhang, Huan
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Gustafsson, Mika
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Benson, Mikael
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center of Paediatrics and Gynaecology and Obstetrics, H.K.H. Kronprinsessan Victorias barn- och ungdomssjukhus.
    Correction: A validated single-cell-based strategy to identify diagnostic and therapeutic targets in complex diseases (vol 11, 47, 2019)2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 37Article in journal (Other academic)
    Abstract [en]

    An amendment to this paper has been published and can be accessed via the original article.

    Download full text (pdf)
    fulltext
  • 14.
    Gawel, Danuta
    et al.
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Serra-Musach, Jordi
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Lilja, Sandra
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Aagesen, Jesper
    Reg Jonkoping Cty, Sweden.
    Arenas, Alex
    Univ Rovira and Virgili, Spain.
    Asking, Bengt
    Reg Jonkoping Cty, Sweden.
    Bengner, Malin
    Reg Jonkoping Cty, Sweden.
    Bjorkander, Janne
    Reg Jonkoping Cty, Sweden.
    Biggs, Sophie
    Linköping University, Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Science. Linköping University, Faculty of Medicine and Health Sciences.
    Ernerudh, Jan
    Linköping University, Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Science. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Diagnostics, Department of Clinical Immunology and Transfusion Medicine.
    Hjortswang, Henrik
    Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Heart and Medicine Center, Department of Gastroentorology. Linköping University, Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Science.
    Karlsson, Jan-Erik
    Linköping University, Department of Medical and Health Sciences, Division of Cardiovascular Medicine. Linköping University, Faculty of Medicine and Health Sciences. Reg Jonkoping Cty, Sweden.
    Köpsén, Mattias
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Jung Lee, Eun Jung
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences. Yonsei Univ, South Korea.
    Lentini, Antonio
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Li, Xinxiu
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Magnusson, Mattias
    Linköping University, Department of Clinical and Experimental Medicine, Division of Neuro and Inflammation Science. Linköping University, Faculty of Medicine and Health Sciences.
    Martinez, David
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Matussek, Andreas
    Reg Jonkoping Cty, Sweden; Karolinska Inst, Sweden; Karolinska Univ Hosp, Sweden.
    Nestor, Colm
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Schäfer, Samuel
    Linköping University, Faculty of Medicine and Health Sciences. Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health.
    Seifert, Oliver
    Linköping University, Department of Clinical and Experimental Medicine, Division of Cell Biology. Linköping University, Faculty of Medicine and Health Sciences. Reg Jonkoping Cty, Sweden.
    Sonmez, Ceylan
    Linköping University, Department of Medical and Health Sciences, Division of Drug Research. Linköping University, Faculty of Medicine and Health Sciences.
    Stjernman, Henrik
    Reg Jonkoping Cty, Sweden.
    Tjärnberg, Andreas
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Wu, Simon
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Åkesson, Karin
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences. Reg Jonkoping Cty, Sweden.
    Shalek, Alex K.
    MIT, MA 02139 USA; Broad Inst MIT and Harvard, MA 02142 USA; Ragon Inst MGH MIT and Harvard, MA USA.
    Stenmarker, Margaretha
    Reg Jonkoping Cty, Sweden; Inst Clin Sci, Sweden.
    Zhang, Huan
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences.
    Gustafsson, Mika
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Benson, Mikael
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center of Paediatrics and Gynaecology and Obstetrics, H.K.H. Kronprinsessan Victorias barn- och ungdomssjukhus Linköping/Motala.
    A validated single-cell-based strategy to identify diagnostic and therapeutic targets in complex diseases2019In: Genome Medicine, E-ISSN 1756-994X, Vol. 11, article id 47Article in journal (Refereed)
    Abstract [en]

    Background

    Genomic medicine has paved the way for identifying biomarkers and therapeutically actionable targets for complex diseases, but is complicated by the involvement of thousands of variably expressed genes across multiple cell types. Single-cell RNA-sequencing study (scRNA-seq) allows the characterization of such complex changes in whole organs.

    Methods

    The study is based on applying network tools to organize and analyze scRNA-seq data from a mouse model of arthritis and human rheumatoid arthritis, in order to find diagnostic biomarkers and therapeutic targets. Diagnostic validation studies were performed using expression profiling data and potential protein biomarkers from prospective clinical studies of 13 diseases. A candidate drug was examined by a treatment study of a mouse model of arthritis, using phenotypic, immunohistochemical, and cellular analyses as read-outs.

    Results

    We performed the first systematic analysis of pathways, potential biomarkers, and drug targets in scRNA-seq data from a complex disease, starting with inflamed joints and lymph nodes from a mouse model of arthritis. We found the involvement of hundreds of pathways, biomarkers, and drug targets that differed greatly between cell types. Analyses of scRNA-seq and GWAS data from human rheumatoid arthritis (RA) supported a similar dispersion of pathogenic mechanisms in different cell types. Thus, systems-level approaches to prioritize biomarkers and drugs are needed. Here, we present a prioritization strategy that is based on constructing network models of disease-associated cell types and interactions using scRNA-seq data from our mouse model of arthritis, as well as human RA, which we term multicellular disease models (MCDMs). We find that the network centrality of MCDM cell types correlates with the enrichment of genes harboring genetic variants associated with RA and thus could potentially be used to prioritize cell types and genes for diagnostics and therapeutics. We validated this hypothesis in a large-scale study of patients with 13 different autoimmune, allergic, infectious, malignant, endocrine, metabolic, and cardiovascular diseases, as well as a therapeutic study of the mouse arthritis model.

    Conclusions

    Overall, our results support that our strategy has the potential to help prioritize diagnostic and therapeutic targets in human disease.

    Download full text (pdf)
    fulltext
  • 15. Gottlieb, Assaf
    et al.
    Daneshjou, Roxana
    DeGorter, M
    Bourgeois, S
    Svensson, PJ
    Wadelius, Mia
    Uppsala University, Science for Life Laboratory, SciLifeLab. Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Sciences, Clinical pharmacogenomics and osteoporosis.
    Deloukas, P
    Montgomery, SB
    Altman, RB
    Cohort-specific imputation of gene expression improves prediction of warfarin dose for African Americans2017In: Genome Medicine, E-ISSN 1756-994X, Vol. 9, no 1, article id 98Article in journal (Refereed)
    Abstract [en]

    BACKGROUND: Genome-wide association studies are useful for discovering genotype-phenotype associations but are limited because they require large cohorts to identify a signal, which can be population-specific. Mapping genetic variation to genes improves power and allows the effects of both protein-coding variation as well as variation in expression to be combined into "gene level" effects.

    METHODS: Previous work has shown that warfarin dose can be predicted using information from genetic variation that affects protein-coding regions. Here, we introduce a method that improves dose prediction by integrating tissue-specific gene expression. In particular, we use drug pathways and expression quantitative trait loci knowledge to impute gene expression-on the assumption that differential expression of key pathway genes may impact dose requirement. We focus on 116 genes from the pharmacokinetic and pharmacodynamic pathways of warfarin within training and validation sets comprising both European and African-descent individuals.

    RESULTS: We build gene-tissue signatures associated with warfarin dose in a cohort-specific manner and identify a signature of 11 gene-tissue pairs that significantly augments the International Warfarin Pharmacogenetics Consortium dosage-prediction algorithm in both populations.

    CONCLUSIONS: Our results demonstrate that imputed expression can improve dose prediction and bridge population-specific compositions.

    MATLAB code is available at https://github.com/assafgo/warfarin-cohort.

    Download full text (pdf)
    fulltext
  • 16. Gudmundsdottir, Valborg
    et al.
    Hong, Mun-Gwan
    KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Thomas, Cecilia Engel
    KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science, Affinity Proteomics. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Schwenk, Jochen M.
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science, Affinity Proteomics.
    Brunak, Soren
    Whole blood co-expression modules associate with metabolic traits and type 2 diabetes: an IMI-DIRECT study2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 109Article in journal (Refereed)
    Abstract [en]

    Background The rising prevalence of type 2 diabetes (T2D) poses a major global challenge. It remains unresolved to what extent transcriptomic signatures of metabolic dysregulation and T2D can be observed in easily accessible tissues such as blood. Additionally, large-scale human studies are required to further our understanding of the putative inflammatory component of insulin resistance and T2D. Here we used transcriptomics data from individuals with (n = 789) and without (n = 2127) T2D from the IMI-DIRECT cohorts to describe the co-expression structure of whole blood that mainly reflects processes and cell types of the immune system, and how it relates to metabolically relevant clinical traits and T2D. Methods Clusters of co-expressed genes were identified in the non-diabetic IMI-DIRECT cohort and evaluated with regard to stability, as well as preservation and rewiring in the cohort of individuals with T2D. We performed functional and immune cell signature enrichment analyses, and a genome-wide association study to describe the genetic regulation of the modules. Phenotypic and trans-omics associations of the transcriptomic modules were investigated across both IMI-DIRECT cohorts. Results We identified 55 whole blood co-expression modules, some of which clustered in larger super-modules. We identified a large number of associations between these transcriptomic modules and measures of insulin action and glucose tolerance. Some of the metabolically linked modules reflect neutrophil-lymphocyte ratio in blood while others are independent of white blood cell estimates, including a module of genes encoding neutrophil granule proteins with antibacterial properties for which the strongest associations with clinical traits and T2D status were observed. Through the integration of genetic and multi-omics data, we provide a holistic view of the regulation and molecular context of whole blood transcriptomic modules. We furthermore identified an overlap between genetic signals for T2D and co-expression modules involved in type II interferon signaling. Conclusions Our results offer a large-scale map of whole blood transcriptomic modules in the context of metabolic disease and point to novel biological candidates for future studies related to T2D.

  • 17. Gudmundsdottir, Valborg
    et al.
    Pedersen, Helle Krogh
    Mazzoni, Gianluca
    Allin, Kristine H.
    Artati, Anna
    Beulens, Joline W.
    Banasik, Karina
    Brorsson, Caroline
    Cederberg, Henna
    Chabanova, Elizaveta
    De Masi, Federico
    Elders, Petra J.
    Forgie, Ian
    Giordano, Giuseppe N.
    Grallert, Harald
    Gupta, Ramneek
    Haid, Mark
    Hansen, Torben
    Hansen, Tue H.
    Hattersley, Andrew T.
    Heggie, Alison
    Hong, Mun-Gwan
    Jones, Angus G.
    Koivula, Robert
    Kokkola, Tarja
    Laakso, Markku
    Longreen, Peter
    Mahajan, Anubha
    Mari, Andrea
    McDonald, Timothy J.
    McEvoy, Donna
    Musholt, Petra B.
    Pavo, Imre
    Prehn, Cornelia
    Ruetten, Hartmut
    Ridderstrale, Martin
    Rutters, Femke
    Sharma, Sapna
    Slieker, Roderick C.
    Syed, Ali
    Tajes, Juan Fernandez
    Thomas, Cecilia Engel
    Thomsen, Henrik S.
    Vangipurapu, Jagadish
    Vestergaard, Henrik
    Vinuela, Ana
    Wesolowska-Andersen, Agata
    Walker, Mark
    Adamski, Jerzy
    Schwenk, Jochen M.
    McCarthy, Mark, I
    Pearson, Ewan
    Dermitzakis, Emmanouil
    Franks, Paul W.
    Umeå University, Faculty of Medicine, Department of Public Health and Clinical Medicine, Section of Medicine. Genetic and Molecular Epidemiology Unit, Lund University Diabetes Centre, Department of Clinical Sciences, Clinical Research Centre, Lund University, Skåne University Hospital, Malmö, Sweden; Oxford Centre for Diabetes, Endocrinology and Metabolism, University of Oxford, Churchill Hospital, Oxford, UK; Department of Nutrition, Harvard School of Public Health, Boston, MA, USA.
    Pedersen, Oluf
    Brunak, Soren
    Whole blood co-expression modules associate with metabolic traits and type 2 diabetes: an IMI-DIRECT study2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 109Article in journal (Refereed)
    Abstract [en]

    Background: The rising prevalence of type 2 diabetes (T2D) poses a major global challenge. It remains unresolved to what extent transcriptomic signatures of metabolic dysregulation and T2D can be observed in easily accessible tissues such as blood. Additionally, large-scale human studies are required to further our understanding of the putative inflammatory component of insulin resistance and T2D. Here we used transcriptomics data from individuals with (n = 789) and without (n = 2127) T2D from the IMI-DIRECT cohorts to describe the co-expression structure of whole blood that mainly reflects processes and cell types of the immune system, and how it relates to metabolically relevant clinical traits and T2D.

    Methods: Clusters of co-expressed genes were identified in the non-diabetic IMI-DIRECT cohort and evaluated with regard to stability, as well as preservation and rewiring in the cohort of individuals with T2D. We performed functional and immune cell signature enrichment analyses, and a genome-wide association study to describe the genetic regulation of the modules. Phenotypic and trans-omics associations of the transcriptomic modules were investigated across both IMI-DIRECT cohorts.

    Results: We identified 55 whole blood co-expression modules, some of which clustered in larger super-modules. We identified a large number of associations between these transcriptomic modules and measures of insulin action and glucose tolerance. Some of the metabolically linked modules reflect neutrophil-lymphocyte ratio in blood while others are independent of white blood cell estimates, including a module of genes encoding neutrophil granule proteins with antibacterial properties for which the strongest associations with clinical traits and T2D status were observed. Through the integration of genetic and multi-omics data, we provide a holistic view of the regulation and molecular context of whole blood transcriptomic modules. We furthermore identified an overlap between genetic signals for T2D and co-expression modules involved in type II interferon signaling.

    Conclusions: Our results offer a large-scale map of whole blood transcriptomic modules in the context of metabolic disease and point to novel biological candidates for future studies related to T2D.

    Download full text (pdf)
    fulltext
  • 18.
    Gustafsson, Mika
    et al.
    The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
    Edström, Måns
    Clinical and Experimental Medicine, Faculty of Health Sciences, Division of Clinical Immunology, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden.
    Gawel, Danuta
    The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
    Nestor, Colm E.
    The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
    Wang, Hui
    The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
    Zhang, Huan
    The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
    Barrenäs, Fredrik
    The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
    Tojo, James
    Department of Clinical Neurosciences, Karolinska Institutet and Centrum for Molecular Medicine, Stockholm, Sweden.
    Kockum, Ingrid
    Department of Clinical Neurosciences, Karolinska Institutet and Centrum for Molecular Medicine, Stockholm, Sweden.
    Olsson, Tomas
    Department of Clinical Neurosciences, Karolinska Institutet and Centrum for Molecular Medicine, Stockholm, Sweden.
    Serra-Musach, Jordi
    Cancer and Systems Biology Unit, Catalan Institute of Oncology, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain.
    Bonifaci, Núria
    Cancer and Systems Biology Unit, Catalan Institute of Oncology, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain.
    Pujana, Miguel
    Cancer and Systems Biology Unit, Catalan Institute of Oncology, IDIBELL, L'Hospitalet del Llobregat, Barcelona, Spain.
    Ernerudh, Jan
    Clinical and Experimental Medicine, Faculty of Health Sciences, Division of Clinical Immunology, Unit of Autoimmunity and Immune Regulation, Linköping University, Linköping, Sweden.
    Benson, Mikael
    The Centre for Individualised Medicine, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.
    Integrated genomic and prospective clinical studies show the importance of modular pleiotropy for disease susceptibility, diagnosis and treatment2014In: Genome Medicine, E-ISSN 1756-994X, Vol. 6, no 2, article id 17Article in journal (Refereed)
    Abstract [en]

    Background: Translational research typically aims to identify and functionally validate individual, disease-specific genes. However, reaching this aim is complicated by the involvement of thousands of genes in common diseases, and that many of those genes are pleiotropic, that is, shared by several diseases.

    Methods: We integrated genomic meta-analyses with prospective clinical studies to systematically investigate the pathogenic, diagnostic and therapeutic roles of pleiotropic genes. In a novel approach, we first used pathway analysis of all published genome-wide association studies (GWAS) to find a cell type common to many diseases.

    Results: The analysis showed over-representation of the T helper cell differentiation pathway, which is expressed in T cells. This led us to focus on expression profiling of CD4(+) T cells from highly diverse inflammatory and malignant diseases. We found that pleiotropic genes were highly interconnected and formed a pleiotropic module, which was enriched for inflammatory, metabolic and proliferative pathways. The general relevance of this module was supported by highly significant enrichment of genetic variants identified by all GWAS and cancer studies, as well as known diagnostic and therapeutic targets. Prospective clinical studies of multiple sclerosis and allergy showed the importance of both pleiotropic and disease specific modules for clinical stratification.

    Conclusions: In summary, this translational genomics study identified a pleiotropic module, which has key pathogenic, diagnostic and therapeutic roles.

  • 19.
    Gustafsson, Mika
    et al.
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Edström, Måns
    Linköping University, Department of Clinical and Experimental Medicine, Division of Inflammation Medicine. Linköping University, Faculty of Health Sciences.
    Gawel, Danuta
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Nestor, Colm
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Wang, Hui
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Zhang, Huan
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Barrenäs, Fredrik
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Tojo, James
    Karolinska Institute, Sweden Centre Molecular Med, Sweden .
    Kockum, Ingrid
    Karolinska Institute, Sweden Centre Molecular Med, Sweden .
    Olsson, Tomas
    Karolinska Institute, Sweden Centre Molecular Med, Sweden .
    Serra-Musach, Jordi
    IDIBELL, Spain .
    Bonifaci, Nuria
    IDIBELL, Spain .
    Angel Pujana, Miguel
    IDIBELL, Spain .
    Ernerudh, Jan
    Linköping University, Department of Clinical and Experimental Medicine, Division of Inflammation Medicine. Linköping University, Faculty of Health Sciences. Östergötlands Läns Landsting, Center for Diagnostics, Department of Clinical Immunology and Transfusion Medicine.
    Benson, Mikael
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences. Östergötlands Läns Landsting, Heart and Medicine Center, Allergy Center. Östergötlands Läns Landsting, Center of Paediatrics and Gynaecology and Obstetrics, Department of Paediatrics in Linköping.
    Integrated genomic and prospective clinical studies show the importance of modular pleiotropy for disease susceptibility, diagnosis and treatment2014In: Genome Medicine, E-ISSN 1756-994X, Vol. 6, no 17Article in journal (Refereed)
    Abstract [en]

    Background: Translational research typically aims to identify and functionally validate individual, disease-specific genes. However, reaching this aim is complicated by the involvement of thousands of genes in common diseases, and that many of those genes are pleiotropic, that is, shared by several diseases. Methods: We integrated genomic meta-analyses with prospective clinical studies to systematically investigate the pathogenic, diagnostic and therapeutic roles of pleiotropic genes. In a novel approach, we first used pathway analysis of all published genome-wide association studies (GWAS) to find a cell type common to many diseases. Results: The analysis showed over-representation of the T helper cell differentiation pathway, which is expressed in T cells. This led us to focus on expression profiling of CD4(+) T cells from highly diverse inflammatory and malignant diseases. We found that pleiotropic genes were highly interconnected and formed a pleiotropic module, which was enriched for inflammatory, metabolic and proliferative pathways. The general relevance of this module was supported by highly significant enrichment of genetic variants identified by all GWAS and cancer studies, as well as known diagnostic and therapeutic targets. Prospective clinical studies of multiple sclerosis and allergy showed the importance of both pleiotropic and disease specific modules for clinical stratification. Conclusions: In summary, this translational genomics study identified a pleiotropic module, which has key pathogenic, diagnostic and therapeutic roles.

    Download full text (pdf)
    fulltext
  • 20.
    Gustafsson, Mika
    et al.
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Nestor, Colm
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Zhang, Huan
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences.
    Barabasi, Albert-Laszlo
    Northeastern University, MA 02115 USA.
    Baranzini, Sergio
    University of Calif San Francisco, CA 94143 USA.
    Brunak, Soeren
    Technical University of Denmark, Denmark; University of Copenhagen, Denmark.
    Fan Chung, Kian
    University of London Imperial Coll Science Technology and Med, England.
    Federoff, Howard J.
    Georgetown University, DC 20057 USA.
    Gavin, Anne-Claude
    European Molecular Biol Lab, Germany.
    Meehan, Richard R.
    University of Edinburgh, Scotland.
    Picotti, Paola
    University of Zurich, Switzerland.
    Angel Pujana, Miguel
    Bellvitge Biomed Research Institute IDIBELL, Spain.
    Rajewsky, Nikolaus
    Max Delbruck Centre Molecular Med, Germany.
    Smith, Kenneth G. C.
    University of Cambridge, England; University of Cambridge, England.
    Sterk, Peter J.
    University of Amsterdam, Netherlands.
    Villoslada, Pablo
    Hospital Clin Barcelona, Spain; Hospital Clin Barcelona, Spain.
    Benson, Mikael
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Health Sciences. Östergötlands Läns Landsting, Heart and Medicine Center, Allergy Center. Östergötlands Läns Landsting, Center of Paediatrics and Gynaecology and Obstetrics, Department of Paediatrics in Linköping.
    Modules, networks and systems medicine for understanding disease and aiding diagnosis2014In: Genome Medicine, E-ISSN 1756-994X, Vol. 6, no 82Article, review/survey (Refereed)
    Abstract [en]

    Many common diseases, such as asthma, diabetes or obesity, involve altered interactions between thousands of genes. High-throughput techniques (omics) allow identification of such genes and their products, but functional understanding is a formidable challenge. Network-based analyses of omics data have identified modules of disease-associated genes that have been used to obtain both a systems level and a molecular understanding of disease mechanisms. For example, in allergy a module was used to find a novel candidate gene that was validated by functional and clinical studies. Such analyses play important roles in systems medicine. This is an emerging discipline that aims to gain a translational understanding of the complex mechanisms underlying common diseases. In this review, we will explain and provide examples of how network-based analyses of omics data, in combination with functional and clinical studies, are aiding our understanding of disease, as well as helping to prioritize diagnostic markers or therapeutic candidate genes. Such analyses involve significant problems and limitations, which will be discussed. We also highlight the steps needed for clinical implementation.

    Download full text (pdf)
    fulltext
  • 21. Howard, Heidi Carmen
    et al.
    Borry, Pascal
    Survey of European clinical geneticists on awareness, experiences and attitudes towards direct-to-consumer genetic testing.2013In: Genome Medicine, E-ISSN 1756-994X, Vol. 5, no 5, p. 45-Article in journal (Refereed)
    Abstract [en]

    BACKGROUND: The advent of direct-to-consumer (DTC) genetic testing (GT) has sparked a number of debates regarding the scientific validity of tests, their broad health and ethical implications for society as well as their legal status. To date, relatively few empirical studies have been published regarding this phenomenon. We conducted a survey of European clinical geneticists to gauge their awareness of, experiences with, and attitudes towards DTC GT.

    METHODS: We invited 300 clinical geneticists from 28 European countries to complete an online questionnaire. Statistical analyses of closed-ended questions were performed using the STATISTICA software package. Answers to open-ended questions were analysed for recurring themes.

    RESULTS: One hundred and thirty-one clinical geneticists answered our survey (response rate, 44%). Eighty-six percent (110/128) of respondents were aware of DTC GT, and over one-third had been contacted by at least one patient regarding these services. The majority (84%) of respondents did not agree with telephone medical supervision outside of an established doctor-patient relationship. The majority of clinical geneticists also found it unacceptable to provide non-face-to-face medical supervision for: (i) a presymptomatic test for a condition with very high penetrance; (ii) a predictive test for a condition that has a 'medium' penetrance of 50% to 60%; and (iii) carrier testing. For conditions that are neither treatable nor preventable and for disorders with serious health consequences, clinical geneticists were almost unanimous in expressing the unacceptability of offering such genetic tests outside of the traditional healthcare setting, without an established physician-patient relationship and without face-to-face medical supervision.

    CONCLUSION: A high percentage of European clinical geneticists are aware of DTC GT and the majority do not agree with the model of provision used by many commercial companies for certain severe or actionable health conditions. Despite this disagreement with the DTC model of provision, >85% of respondents said that they would offer genetic counselling to patients who asked for a consultation after having undergone DTC genetic testing. The understanding of the views and opinions of this expert stakeholder group should be considered in the attempts to shape responsible policy and guidelines for these services.

  • 22.
    Kundu, Snehangshu
    et al.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology. Uppsala University, Science for Life Laboratory, SciLifeLab.
    Ali, Muhammad Akhtar
    Uppsala University, Science for Life Laboratory, SciLifeLab. Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology.
    Handin, Niklas
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Pharmacy, Department of Pharmacy.
    Padhan, Narendra
    Uppsala University, Science for Life Laboratory, SciLifeLab. Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Vascular Biology.
    Larsson, Jimmy
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology. Uppsala University, Science for Life Laboratory, SciLifeLab.
    Karoutsou, Maria
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology. Uppsala University, Science for Life Laboratory, SciLifeLab.
    Ban, Kenneth
    Natl Univ Singapore, Yong Loo Lin Sch Med, Dept Biochem, 8 Med Dr,02-06, Singapore 117597, Singapore.;ASTAR, Inst Mol & Cell Biol, Singapore 138673, Singapore..
    Wisniewski, Jacek R.
    Max Planck Inst Biochem, Dept Prote & Signal Transduct, Biochem Prote Grp, D-82152 Martinsried, Germany..
    Artursson, Per
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Pharmacy, Department of Pharmacy.
    He, Liqun
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Vascular Biology. Uppsala University, Science for Life Laboratory, SciLifeLab. Department of Neurosurgery, Tianjin Medical University General Hospital, Tianjin Neurological Institute, Key Laboratory of Post-Neuroinjury Neuro-Repair and Regeneration in Central Nervous System, Ministry of Education and Tianjin City, Tianjin, China.
    Hellström, Mats
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology. Uppsala University, Science for Life Laboratory, SciLifeLab.
    Sjöblom, Tobias
    Uppsala University, Science for Life Laboratory, SciLifeLab. Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Immunology, Genetics and Pathology, Experimental and Clinical Oncology.
    Linking FOXO3, NCOA3, and TCF7L2 to Ras pathway phenotypes through a genome-wide forward genetic screen in human colorectal cancer cells2018In: Genome Medicine, E-ISSN 1756-994X, Vol. 10, article id 2Article in journal (Refereed)
    Abstract [en]

    Background:

    The Ras pathway genes KRAS, BRAF, or ERBBs have somatic mutations in similar to 60% of human colorectal carcinomas. At present, it is unknown whether the remaining cases lack mutations activating the Ras pathway or whether they have acquired mutations in genes hitherto unknown to belong to the pathway.

    Methods:

    To address the second possibility and extend the compendium of Ras pathway genes, we used genome-wide transposon mutagenesis of two human colorectal cancer cell systems deprived of their activating KRAS or BRAF allele to identify genes enabling growth in low glucose, a Ras pathway phenotype, when targeted.

    Results:

    Of the 163 recurrently targeted genes in the two different genetic backgrounds, one-third were known cancer genes and one-fifth had links to the EGFR/Ras/MAPK pathway. When compared to cancer genome sequencing datasets, nine genes also mutated in human colorectal cancers were identified. Among these, stable knockdown of FOXO3, NCOA3, and TCF7L2 restored growth in low glucose but reduced MEK/MAPK phosphorylation, reduced anchorage-independent growth, and modulated expressions of GLUT1 and Ras pathway related proteins. Knockdown of NCOA3 and FOXO3 significantly decreased the sensitivity to cetuximab of KRAS mutant but not wild-type cells.

    Conclusions:

    This work establishes a proof-of-concept that human cell-based genome-wide forward genetic screens can assign genes to pathways with clinical importance in human colorectal cancer.

  • 23. Larsson, Erik
    et al.
    Fredlund Fuchs, Peder
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Biochemistry and Microbiology.
    Heldin, Johan
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Biochemistry and Microbiology.
    Barkefors, Irmeli
    Uppsala University, Disciplinary Domain of Science and Technology, Biology, Department of Cell and Molecular Biology, Computational and Systems Biology.
    Bondjers, Cecilia
    Genové, Guillem
    Arrondel, Christelle
    Gerwins, Pär
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Biochemistry and Microbiology.
    Kurschat, Christine
    Schermer, Bernhard
    Benzing, Thomas
    Harvey, Scott J
    Kreuger, Johan
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Biochemistry and Microbiology.
    Lindahl, Per
    Discovery of microvascular miRNAs using public gene expression data: miR-145 is expressed in pericytes and is a regulator of Fli12009In: Genome Medicine, E-ISSN 1756-994X, Vol. 1, no 11, p. 108-Article in journal (Refereed)
    Abstract [en]

    BACKGROUND

    A function for the microRNA (miRNA) pathway in vascular development and angiogenesis has been firmly established. miRNAs with selective expression in the vasculature are attractive as possible targets in miRNA-based therapies. However, little is known about the expression of miRNAs in microvessels in vivo. Here, we identified candidate microvascular-selective miRNAs by screening public miRNA expression datasets.

    METHODS

    Bioinformatics predictions of microvascular-selective expression were validated with real-time quantitative reverse transcription PCR on purified microvascular fragments from mouse. Pericyte expression was shown with in situ hybridization on tissue sections. Target sites were identified with 3' UTR luciferase assays, and migration was tested in a microfluid chemotaxis chamber.

    RESULTS

    miR-145, miR-126, miR-24, and miR-23a were selectively expressed in microvascular fragments isolated from a range of tissues. In situ hybridization and analysis of Pdgfb retention motif mutant mice demonstrated predominant expression of miR-145 in pericytes. We identified the Ets transcription factor Friend leukemia virus integration 1 (Fli1) as a miR-145 target, and showed that elevated levels of miR-145 reduced migration of microvascular cells in response to growth factor gradients in vitro.

    CONCLUSIONS

    miR-126, miR-24 and miR-23a are selectively expressed in microvascular endothelial cells in vivo, whereas miR-145 is expressed in pericytes. miR-145 targets the hematopoietic transcription factor Fli1 and blocks migration in response to growth factor gradients. Our findings have implications for vascular disease and provide necessary information for future drug design against miRNAs with selective expression in the microvasculature.

  • 24.
    Li, Xinxiu
    et al.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Jung Lee, Eun Jung
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences. Yonsei Univ, South Korea.
    Lilja, Sandra
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Loscalzo, Joseph
    Brigham & Womens Hosp, MA 02115 USA; Harvard Med Sch, MA 02115 USA; Harvard Med Sch, MA 02115 USA.
    Schäfer, Samuel
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Smelik, Martin
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Strobl, Maria Regina
    Med Univ Vienna, Austria.
    Sysoev, Oleg
    Linköping University, Department of Computer and Information Science, The Division of Statistics and Machine Learning. Linköping University, Faculty of Arts and Sciences.
    Wang, Hui
    Xuzhou Med Univ, Peoples R China.
    Zhang, Huan
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Zhao, Yelin
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Gawel, Danuta
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Bohle, Barbara
    Med Univ Vienna, Austria.
    Benson, Mikael
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center of Paediatrics and Gynaecology and Obstetrics, H.K.H. Kronprinsessan Victorias barn- och ungdomssjukhus. Karolinska Inst, Sweden.
    A dynamic single cell-based framework for digital twins to prioritize disease genes and drug targets2022In: Genome Medicine, E-ISSN 1756-994X, Vol. 14, no 1, article id 48Article in journal (Refereed)
    Abstract [en]

    Background Medical digital twins are computational disease models for drug discovery and treatment. Unresolved problems include how to organize and prioritize between disease-associated changes in digital twins, on cellulome- and genome-wide scales. We present a dynamic framework that can be used to model such changes and thereby prioritize upstream regulators (URs) for biomarker- and drug discovery. Methods We started with seasonal allergic rhinitis (SAR) as a disease model, by analyses of in vitro allergen-stimulated peripheral blood mononuclear cells (PBMC) from SAR patients. Time-series a single-cell RNA-sequencing (scRNA-seq) data of these cells were used to construct multicellular network models (MNMs) at each time point of molecular interactions between cell types. We hypothesized that predicted molecular interactions between cell types in the MNMs could be traced to find an UR gene, at an early time point. We performed bioinformatic and functional studies of the MNMs to develop a scalable framework to prioritize UR genes. This framework was tested on a single-cell and bulk-profiling data from SAR and other inflammatory diseases. Results Our scRNA-seq-based time-series MNMs of SAR showed thousands of differentially expressed genes (DEGs) across multiple cell types, which varied between time points. Instead of a single-UR gene in each MNM, we found multiple URs dispersed across the cell types. Thus, at each time point, the MNMs formed multi-directional networks. The absence of linear hierarchies and time-dependent variations in MNMs complicated the prioritization of URs. For example, the expression and functions of Th2 cytokines, which are approved drug targets in allergies, varied across cell types, and time points. Our analyses of bulk- and single-cell data from other inflammatory diseases also revealed multi-directional networks that showed stage-dependent variations. We therefore developed a quantitative approach to prioritize URs: we ranked the URs based on their predicted effects on downstream target cells. Experimental and bioinformatic analyses supported that this kind of ranking is a tractable approach for prioritizing URs. Conclusions We present a scalable framework for modeling dynamic changes in digital twins, on cellulome- and genome-wide scales, to prioritize UR genes for biomarker and drug discovery.

    Download full text (pdf)
    fulltext
  • 25.
    Lindstrand, Anna
    et al.
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Eisfeldt, Jesper
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden.;Karolinska Inst, Sci Life Lab, Stockholm, Sweden..
    Pettersson, Maria
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Carvalho, Claudia M. B.
    Baylor Coll Med, Dept Mol & Human Genet, Houston, TX 77030 USA..
    Kvarnung, Malin
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Grigelioniene, Giedre
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Anderlid, Britt-Marie
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Bjerin, Olof
    Karolinska Inst, Dept Womens & Childrens Hlth, Stockholm, Sweden..
    Gustavsson, Peter
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Hammarsjö, Anna
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Georgii-Hemming, Patrik
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden..
    Iwarsson, Erik
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Johansson-Soller, Maria
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Lagerstedt-Robinson, Kristina
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Lieden, Agne
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Magnusson, Mans
    Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Sci Life Lab, Stockholm, Sweden.;Karolinska Univ Hosp, Ctr Inherited Metab Dis, Stockholm, Sweden..
    Martin, Marcel
    Stockholm Univ, Dept Biochem & Biophys, Sci Life Lab, Natl Bioinformat Infrastruct Sweden, Stockholm, Sweden..
    Malmgren, Helena
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Nordenskjöld, Magnus
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Norling, Ameli
    Karolinska Inst, Dept Womens & Childrens Hlth, Stockholm, Sweden..
    Sahlin, Ellika
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Stranneheim, Henrik
    Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden.;Karolinska Univ Hosp, Ctr Inherited Metab Dis, Stockholm, Sweden..
    Tham, Emma
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Wincent, Josephine
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Ygberg, Sofia
    Karolinska Univ Hosp, Ctr Inherited Metab Dis, Stockholm, Sweden..
    Wedell, Anna
    Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Univ Hosp, Ctr Inherited Metab Dis, Stockholm, Sweden..
    Wirta, Valtteri
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Gene Technology.
    Nordgren, Ann
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden..
    Lundin, Johanna
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden.;Baylor Coll Med, Dept Mol & Human Genet, Houston, TX 77030 USA..
    Nilsson, Daniel
    Karolinska Univ Hosp, Dept Clin Genet, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Inst, Ctr Mol Med, Stockholm, Sweden.;Karolinska Inst, Sci Life Lab, Stockholm, Sweden..
    From cytogenetics to cytogenomics: whole-genome sequencing as a first-line test comprehensively captures the diverse spectrum of disease-causing genetic variation underlying intellectual disability2019In: Genome Medicine, E-ISSN 1756-994X, Vol. 11, no 1, article id 68Article in journal (Refereed)
    Abstract [en]

    BackgroundSince different types of genetic variants, from single nucleotide variants (SNVs) to large chromosomal rearrangements, underlie intellectual disability, we evaluated the use of whole-genome sequencing (WGS) rather than chromosomal microarray analysis (CMA) as a first-line genetic diagnostic test.MethodsWe analyzed three cohorts with short-read WGS: (i) a retrospective cohort with validated copy number variants (CNVs) (cohort 1, n=68), (ii) individuals referred for monogenic multi-gene panels (cohort 2, n=156), and (iii) 100 prospective, consecutive cases referred to our center for CMA (cohort 3). Bioinformatic tools developed include FindSV, SVDB, Rhocall, Rhoviz, and vcf2cytosure.ResultsFirst, we validated our structural variant (SV)-calling pipeline on cohort 1, consisting of three trisomies and 79 deletions and duplications with a median size of 850kb (min 500bp, max 155Mb). All variants were detected. Second, we utilized the same pipeline in cohort 2 and analyzed with monogenic WGS panels, increasing the diagnostic yield to 8%. Next, cohort 3 was analyzed by both CMA and WGS. The WGS data was processed for large (>10kb) SVs genome-wide and for exonic SVs and SNVs in a panel of 887 genes linked to intellectual disability as well as genes matched to patient-specific Human Phenotype Ontology (HPO) phenotypes. This yielded a total of 25 pathogenic variants (SNVs or SVs), of which 12 were detected by CMA as well. We also applied short tandem repeat (STR) expansion detection and discovered one pathologic expansion in ATXN7. Finally, a case of Prader-Willi syndrome with uniparental disomy (UPD) was validated in the WGS data.Important positional information was obtained in all cohorts. Remarkably, 7% of the analyzed cases harbored complex structural variants, as exemplified by a ring chromosome and two duplications found to be an insertional translocation and part of a cryptic unbalanced translocation, respectively.ConclusionThe overall diagnostic rate of 27% was more than doubled compared to clinical microarray (12%). Using WGS, we detected a wide range of SVs with high accuracy. Since the WGS data also allowed for analysis of SNVs, UPD, and STRs, it represents a powerful comprehensive genetic test in a clinical diagnostic laboratory setting.

  • 26. Lindstrand, Anna
    et al.
    Eisfeldt, Jesper
    Pettersson, Maria
    Carvalho, Claudia M. B.
    Kvarnung, Malin
    Grigelioniene, Giedre
    Anderlid, Britt-Marie
    Bjerin, Olof
    Gustavsson, Peter
    Hammarsjö, Anna
    Georgii-Hemming, Patrik
    Iwarsson, Erik
    Johansson-Soller, Maria
    Lagerstedt-Robinson, Kristina
    Lieden, Agne
    Magnusson, Måns
    Martin, Marcel
    Stockholm University, Faculty of Science, Department of Biochemistry and Biophysics. Stockholm University, Science for Life Laboratory (SciLifeLab).
    Malmgren, Helena
    Nordenskjöld, Magnus
    Norling, Ameli
    Sahlin, Ellika
    Stranneheim, Henrik
    Tham, Emma
    Wincent, Josephine
    Ygberg, Sofia
    Wedell, Anna
    Wirta, Valtteri
    Nordgren, Ann
    Lundin, Johanna
    Nilsson, Daniel
    From cytogenetics to cytogenomics: whole-genome sequencing as a first-line test comprehensively captures the diverse spectrum of disease-causing genetic variation underlying intellectual disability2019In: Genome Medicine, E-ISSN 1756-994X, Vol. 11, no 1, article id 68Article in journal (Refereed)
    Abstract [en]

    Background: Since different types of genetic variants, from single nucleotide variants (SNVs) to large chromosomal rearrangements, underlie intellectual disability, we evaluated the use of whole-genome sequencing (WGS) rather than chromosomal microarray analysis (CMA) as a first-line genetic diagnostic test.

    Methods: We analyzed three cohorts with short-read WGS: (i) a retrospective cohort with validated copy number variants (CNVs) (cohort 1, n=68), (ii) individuals referred for monogenic multi-gene panels (cohort 2, n=156), and (iii) 100 prospective, consecutive cases referred to our center for CMA (cohort 3). Bioinformatic tools developed include FindSV, SVDB, Rhocall, Rhoviz, and vcf2cytosure.

    Results: First, we validated our structural variant (SV)-calling pipeline on cohort 1, consisting of three trisomies and 79 deletions and duplications with a median size of 850kb (min 500bp, max 155Mb). All variants were detected. Second, we utilized the same pipeline in cohort 2 and analyzed with monogenic WGS panels, increasing the diagnostic yield to 8%. Next, cohort 3 was analyzed by both CMA and WGS. The WGS data was processed for large (>10kb) SVs genome-wide and for exonic SVs and SNVs in a panel of 887 genes linked to intellectual disability as well as genes matched to patient-specific Human Phenotype Ontology (HPO) phenotypes. This yielded a total of 25 pathogenic variants (SNVs or SVs), of which 12 were detected by CMA as well. We also applied short tandem repeat (STR) expansion detection and discovered one pathologic expansion in ATXN7. Finally, a case of Prader-Willi syndrome with uniparental disomy (UPD) was validated in the WGS data. Important positional information was obtained in all cohorts. Remarkably, 7% of the analyzed cases harbored complex structural variants, as exemplified by a ring chromosome and two duplications found to be an insertional translocation and part of a cryptic unbalanced translocation, respectively.

    Conclusion: The overall diagnostic rate of 27% was more than doubled compared to clinical microarray (12%). Using WGS, we detected a wide range of SVs with high accuracy. Since the WGS data also allowed for analysis of SNVs, UPD, and STRs, it represents a powerful comprehensive genetic test in a clinical diagnostic laboratory setting.

  • 27.
    Merker, Matthias
    et al.
    German Ctr Infect Res DZIF, Germany; Res Ctr Borstel, Germany.
    Kohl, Thomas A.
    German Ctr Infect Res DZIF, Germany; Res Ctr Borstel, Germany.
    Barilar, Ivan
    German Ctr Infect Res DZIF, Germany; Res Ctr Borstel, Germany.
    Andres, Soenke
    Natl and WHO Supranat Reference Ctr Mycobacteria, Germany.
    Fowler, Philip W.
    Univ Oxford, England.
    Chryssanthou, Erja
    Karolinska Univ Hosp, Sweden; Karolinska Inst, Sweden.
    Angeby, Kristian
    Karolinska Inst, Sweden.
    Jureen, Pontus
    Publ Hlth Agcy Sweden, Sweden.
    Moradigaravand, Danesh
    Univ Birmingham, England.
    Parkhill, Julian
    Univ Cambridge, England.
    Peacock, Sharon J.
    Univ Cambridge, England.
    Schön, Thomas
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection. Linköping University, Faculty of Medicine and Health Sciences. Kalmar Cty Hosp, Sweden.
    Maurer, Florian P.
    Natl and WHO Supranat Reference Ctr Mycobacteria, Germany; Univ Med Ctr Hamburg Eppendorf, Germany.
    Walker, Timothy
    Univ Oxford, England.
    Koser, Claudio
    Univ Cambridge, England.
    Niemann, Stefan
    German Ctr Infect Res DZIF, Germany; Res Ctr Borstel, Germany.
    Phylogenetically informative mutations in genes implicated in antibiotic resistance in Mycobacterium tuberculosis complex2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 27Article in journal (Refereed)
    Abstract [en]

    Background A comprehensive understanding of the pre-existing genetic variation in genes associated with antibiotic resistance in the Mycobacterium tuberculosis complex (MTBC) is needed to accurately interpret whole-genome sequencing data for genotypic drug susceptibility testing (DST). Methods We investigated mutations in 92 genes implicated in resistance to 21 anti-tuberculosis drugs using the genomes of 405 phylogenetically diverse MTBC strains. The role of phylogenetically informative mutations was assessed by routine phenotypic DST data for the first-line drugs isoniazid, rifampicin, ethambutol, and pyrazinamide from a separate collection of over 7000 clinical strains. Selected mutations/strains were further investigated by minimum inhibitory concentration (MIC) testing. Results Out of 547 phylogenetically informative mutations identified, 138 were classified as not correlating with resistance to first-line drugs. MIC testing did not reveal a discernible impact of a Rv1979c deletion shared by M. africanum lineage 5 strains on resistance to clofazimine. Finally, we found molecular evidence that some MTBC subgroups may be hyper-susceptible to bedaquiline and clofazimine by different loss-of-function mutations affecting a drug efflux pump subunit (MmpL5). Conclusions Our findings underline that the genetic diversity in MTBC has to be studied more systematically to inform the design of clinical trials and to define sound epidemiologic cut-off values (ECOFFs) for new and repurposed anti-tuberculosis drugs. In that regard, our comprehensive variant catalogue provides a solid basis for the interpretation of mutations in genotypic as well as in phenotypic DST assays.

    Download full text (pdf)
    fulltext
  • 28.
    Mignardi, Marco
    et al.
    Stockholm University, Faculty of Science, Department of Biochemistry and Biophysics. Stockholm University, Science for Life Laboratory (SciLifeLab).
    Nilsson, Mats
    Stockholm University, Faculty of Science, Department of Biochemistry and Biophysics. Stockholm University, Science for Life Laboratory (SciLifeLab).
    Fourth-generation sequencing in the cell and the clinic2014In: Genome Medicine, E-ISSN 1756-994X, Vol. 6, p. 31-31Article in journal (Other academic)
    Abstract [en]

    Nearly 40 years ago, DNA was sequenced for the first time. Since then, DNA sequencing has undergone continuous development, passing through three generations of sequencing technology. We are now entering the beginning of a new phase of genomic analysis in which massively parallel sequencing is performed directly in the cell. Two methods have recently been described for in situ RNA sequencing, one targeted and one untargeted, that rely on ligation chemistry. This fourth generation of sequencing technology opens up prospects for transcriptomic analysis, biomarker validation, diagnosis and patient stratification for cancer treatment.

  • 29.
    Milne, Richard
    et al.
    Wellcome Genome Campus, Wellcome Connecting Sci, Soc & Eth Res Grp, Cambridge CB10 1SA, England; Univ Cambridge, Dept Publ Hlth & Primary Care, Cambridge CB2 0SR, England.
    Morley, Katherine I.
    RAND Europe, Cambridge CB4 1YG, England; Kings Coll London, Inst Psychiat Psychol & Neurosci, London SE5 8AF, England; Univ Melbourne, Melbourne Sch Global & Populat Hlth, Ctr Epidemiol & Biostat, Melbourne, Vic 3010, Australia.
    Almarri, Mohamed A.
    Wellcome Sanger Inst, Cambridge CB10 1SA, England; Dubai Police GHQ, Dept Forens Sci & Criminol, Dubai, U Arab Emirates.
    Anwer, Shamim
    Keynote IAS, New Delhi 110060, India.
    Atutornu, Jerome
    Wellcome Genome Campus, Wellcome Connecting Sci, Soc & Eth Res Grp, Cambridge CB10 1SA, England.
    Baranova, Elena E.
    Russian Med Acad Continuous Profess Educ, Moscow 119049, Russia.
    Bevan, Paul
    Wellcome Sanger Inst, Cambridge CB10 1SA, England.
    Cerezo, Maria
    Wellcome Genome Campus, EMBL EBI, Cambridge CB10 1SA, England.
    Cong, Yali
    Peking Univ, Med Eth Program, Hlth Sci Ctr, Beijing 100191, Peoples R China.
    Costa, Alessia
    Wellcome Genome Campus, Wellcome Connecting Sci, Soc & Eth Res Grp, Cambridge CB10 1SA, England.
    Critchley, Christine
    Swinburne Univ Technol, Dept Psychol Sci, Melbourne, Vic 3122, Australia; Univ Tasmania, Ctr Law & Genet, Hobart, Tas 7001, Australia.
    Fernow, Josepine
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics.
    Goodhand, Peter
    MaRS Ctr, Ontario Inst Canc Res, Toronto, ON M5G 0A3, Canada.
    Hasan, Qurratulain
    Kamineni Hosp, Dept Genet & Mol Med, Hyderabad 500068, India; SAAZ Genet, Hyderabad 500033, India.
    Hibino, Aiko
    Hirosaki Univ, Fac Humanities & Social Sci, Hirosaki, Aomori 0368560, Japan.
    Houeland, Gry
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics.
    Howard, Heidi C.
    Lund Univ, Med Eth, Sölvegatan 19, Lund, Sweden.
    Hussain, S. Zakir
    SAAZ Genet, Hyderabad 500033, India.
    Ingvoldstad Malmgren, Charlotta
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics. Karolinska Inst, Dept Mol Med & Surg, S-17176 Solna, Sweden.
    Izhevskaya, Vera L.
    Res Ctr Med Genet, Moscow 115522, Russia.
    Jedrzejak, Aleksandra
    Jinhong, Cao
    Wuhan Univ, Sch Hlth Sci, Dept Epidemiol & Biostat, Wuhan 430071, Peoples R China.
    Kimura, Megumi
    Hitotsubashi Univ, Inst Innovat Res, Tokyo 1868603, Japan.
    Kleiderman, Erika
    McGill Univ, Ctr Genom & Policy, Montreal, PQ H3A 0G1, Canada.
    Leach, Brandi
    RAND Europe, Cambridge CB4 1YG, England.
    Liu, Keying
    Osaka Univ, Dept Social Med, Publ Hlth, Grad Sch Med, Osaka 5650871, Japan; Peking Univ, Sch Publ Hlth, Hlth Sci Ctr, Beijing 100191, Peoples R China.
    Mascalzoni, Deborah
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics. EURAC, Inst Biomed, I-39100 Bolzano, Italy.
    Mendes, Álvaro
    Univ Porto, UnIGENe, P-4200135 Porto, Portugal; Univ Porto, CGPP Ctr Predict & Prevent Genet, IBMC Inst Mol & Cell Biol, i3S Inst Invest & Inovacao Saude, P-4200135 Porto, Portugal.
    Minari, Jusaku
    Kyoto Univ, Uehiro Res Div iPS Cell Eth, Ctr iPS Cell Res & Applicat CiRA, Kyoto 6068507, Japan.
    Nicol, Dianne
    Univ Tasmania, Ctr Law & Genet, Hobart, Tas 7001, Australia.
    Niemiec, Emilia
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics.
    Patch, Christine
    Wellcome Genome Campus, Wellcome Connecting Sci, Soc & Eth Res Grp, Cambridge CB10 1SA, England; Queen Mary Univ London, Genom England, London EC1M 6BQ, England.
    Pollard, Jack
    RAND Europe, Cambridge CB4 1YG, England.
    Prainsack, Barbara
    Univ Vienna, Dept Polit Sci, A-1010 Vienna, Austria; Kings Coll London, Dept Global Hlth & Social Med, London WC2R 2LS, England.
    Rivière, Marie
    Sorbonne Nouvelle, DILTEC, F-75005 Paris, France.
    Robarts, Lauren
    Wellcome Genome Campus, Wellcome Connecting Sci, Soc & Eth Res Grp, Cambridge CB10 1SA, England.
    Roberts, Jonathan
    Wellcome Genome Campus, Wellcome Connecting Sci, Soc & Eth Res Grp, Cambridge CB10 1SA, England.
    Romano, Virginia
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics. EURAC, Inst Biomed, I-39100 Bolzano, Italy.
    Sheerah, Haytham A.
    Osaka Univ, Dept Social Med, Publ Hlth, Grad Sch Med, Osaka 5650871, Japan.
    Smith, James
    Wellcome Sanger Inst, Cambridge CB10 1SA, England.
    Soulier, Alexandra
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics.
    Steed, Claire
    Wellcome Sanger Inst, Cambridge CB10 1SA, England.
    Stefansdottir, Vigdis
    Natl Univ Hosp Iceland, Landspitali, IS-101 Reykjavik, Iceland.
    Tandre, Cornelia
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Public Health and Caring Sciences, Centre for Research Ethics and Bioethics.
    Thorogood, Adrian
    McGill Univ, Ctr Genom & Policy, Montreal, PQ H3A 0G1, Canada.
    Voigt, Torsten H.
    Rhein Westfal TH Aachen, Inst Sociol, D-52062 Aachen, Germany.
    Wang, Nan
    Peking Univ, Med Eth Program, Hlth Sci Ctr, Beijing 100191, Peoples R China.
    West, Anne V.
    Indiana Univ, Maurer Sch Law, Bloomington, IN 47405 USA.
    Yoshizawa, Go
    Oslo Metropolitan Univ, Work Res Inst AFI, N-0130 Oslo, Norway.
    Middleton, Anna
    Wellcome Genome Campus, Wellcome Connecting Sci, Soc & Eth Res Grp, Cambridge CB10 1SA, England; Univ Cambridge, Fac Educ, Cambridge CB2 8PQ, England.
    Demonstrating trustworthiness when collecting and sharing genomic data: public views across 22 countries2021In: Genome Medicine, E-ISSN 1756-994X, Vol. 13, no 1, article id 92Article in journal (Refereed)
    Abstract [en]

    Background

    Public trust is central to the collection of genomic and health data and the sustainability of genomic research. To merit trust, those involved in collecting and sharing data need to demonstrate they are trustworthy. However, it is unclear what measures are most likely to demonstrate this.

    Methods

    We analyse the ‘Your DNA, Your Say’ online survey of public perspectives on genomic data sharing including responses from 36,268 individuals across 22 low-, middle- and high-income countries, gathered in 15 languages. We examine how participants perceived the relative value of measures to demonstrate the trustworthiness of those using donated DNA and/or medical information. We examine between-country variation and present a consolidated ranking of measures.

    Results

    Providing transparent information about who will benefit from data access was the most important measure to increase trust, endorsed by more than 50% of participants across 20 of 22 countries. It was followed by the option to withdraw data and transparency about who is using data and why. Variation was found for the importance of measures, notably information about sanctions for misuse of data—endorsed by 5% in India but almost 60% in Japan. A clustering analysis suggests alignment between some countries in the assessment of specific measures, such as the UK and Canada, Spain and Mexico and Portugal and Brazil. China and Russia are less closely aligned with other countries in terms of the value of the measures presented.

    Conclusions

    Our findings highlight the importance of transparency about data use and about the goals and potential benefits associated with data sharing, including to whom such benefits accrue. They show that members of the public value knowing what benefits accrue from the use of data. The study highlights the importance of locally sensitive measures to increase trust as genomic data sharing continues globally.

    Download full text (pdf)
    FULLTEXT01
  • 30.
    Miotto, Paolo
    et al.
    IRCCS San Raffaele Sci Inst, Italy.
    Cirillo, Daniela M.
    IRCCS San Raffaele Sci Inst, Italy.
    Schön, Thomas
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Inflammation and Infection. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Diagnostics, Department of Clinical Microbiology. Region Östergötland, Medicine Center, Department of Infectious Diseases.
    Koeser, Claudio U.
    Univ Cambridge, England.
    The exceptions that prove the rule-a historical view of bedaquiline susceptibility2024In: Genome Medicine, E-ISSN 1756-994X, Vol. 16, no 1, article id 39Article in journal (Refereed)
    Abstract [en]

    In the accompanying study, Nimmo and colleagues estimated the dates of emergence of mutations in mmpR5 (Rv0678), the most important resistance gene to the anti-tuberculosis drug bedaquiline, in over 3500 geographically diverse Mycobacterium tuberculosis genomes. This provided important insights to improve the design and analysis of clinical trials, as well as the World Health Organization catalogue of resistance mutations, the global reference for interpreting genotypic antimicrobial susceptibility testing results.

  • 31.
    Mueller, Stefanie H.
    et al.
    UCL, Inst Hlth Informat, London, England..
    Lai, Alvina G.
    UCL, Inst Hlth Informat, London, England..
    Valkovskaya, Maria
    UCL, Div Psychiat, London, England..
    Michailidou, Kyriaki
    Cyprus Inst Neurol & Genet, Biostat Unit, CY-2371 Nicosia, Cyprus.;Cyprus Inst Neurol & Genet, Cyprus Sch Mol Med, CY-2371 Nicosia, Cyprus.;Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Publ Hlth & Primary Care, Cambridge CB1 8RN, England..
    Bolla, Manjeet K.
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Publ Hlth & Primary Care, Cambridge CB1 8RN, England..
    Wang, Qin
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Publ Hlth & Primary Care, Cambridge CB1 8RN, England..
    Dennis, Joe
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Publ Hlth & Primary Care, Cambridge CB1 8RN, England..
    Lush, Michael
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Publ Hlth & Primary Care, Cambridge CB1 8RN, England..
    Abu-Ful, Zomoruda
    Carmel Hosp, Clalit Natl Canc Control Ctr, IL-35254 Haifa, Israel.;Technion Fac Med, IL-35254 Haifa, Israel..
    Ahearn, Thomas U.
    NCI, Div Canc Epidemiol & Genet, US Dept HHS, NIH, Bethesda, MD 20850 USA..
    Andrulis, Irene L.
    Lunenfeld Tanenbaum Res Inst Mt Sinai Hosp, Fred A Litwin Ctr Canc Genet, Toronto, ON M5G 1X5, Canada.;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 1A8, Canada..
    Anton-Culver, Hoda
    Univ Calif Irvine, Genet Epidemiol Res Inst, Dept Med, Irvine, CA 92617 USA..
    Antonenkova, Natalia N.
    NN Alexandrov Res Inst Oncol & Med Radiol, Minsk 223040, BELARUS..
    Arndt, Volker
    German Canc Res Ctr, Div Clin Epidemiol & Aging Res, D-69120 Heidelberg, Germany..
    Aronson, Kristan J.
    Queens Univ, Canc Res Inst, Dept Publ Hlth Sci, Kingston, ON K7L 3N6, Canada..
    Augustinsson, Annelie
    Lund Univ, Dept Canc Epidemiol, Clin Sci, S-22242 Lund, Sweden..
    Baert, Thais
    Univ Hosp Leuven, Leuven Multidisciplinary Breast Ctr, Leuven Canc Inst, Dept Oncol, B-3000 Louvain, Belgium..
    Freeman, Laura E. Beane
    NCI, Div Canc Epidemiol & Genet, US Dept HHS, NIH, Bethesda, MD 20850 USA..
    Beckmann, Matthias W.
    Friedrich Alexander Univ Erlangen Nuremberg FAU, Univ Hosp Erlangen, Comprehens Canc Ctr Erlangen EMN, Dept Gynecol & Obstet, D-91054 Erlangen, Germany..
    Behrens, Sabine
    German Canc Res Ctr, Div Canc Epidemiol, D-69120 Heidelberg, Germany..
    Benitez, Javier
    Biomed Network Rare Dis CIBERER, Madrid 28029, Spain.;Spanish Natl Canc Res Ctr CNIO, Human Canc Genet Programme, Madrid 28029, Spain..
    Bermisheva, Marina
    Russian Acad Sci, Inst Biochem & Genet, Ufa Fed Res Ctr, Ufa 450054, Russia..
    Blomqvist, Carl
    Univ Helsinki, Helsinki Univ Hosp, Dept Oncol, Helsinki 00290, Finland.;Örebro Univ Hosp, Dept Oncol, S-70185 Örebro, Sweden..
    Bogdanova, Natalia, V
    NN Alexandrov Res Inst Oncol & Med Radiol, Minsk 223040, BELARUS.;Hannover Med Sch, Depart Ment Radiat Oncol, D-30625 Hannover, Germany.;Hannover Med Sch, Gynaecol Res Unit, D-30625 Hannover, Germany..
    Bojesen, Stig E.
    Copenhagen Univ Hosp, Herlev & Gentofte Hosp, Copenhagen Gen Populat Study, DK-2730 Herlev, Denmark.;Copenhagen Univ Hosp, Herlev & Gentofte Hosp, Dept Clin Biochem, DK-2730 Herlev, Denmark.;Univ Copenhagen, Fac Hlth & Med Sci, DK-2200 Copenhagen, Denmark..
    Bonanni, Bernardo
    European Inst Oncol IRCCS, Div Canc Prevent & Genet, IEO, I-20141 Milan, Italy..
    Brenner, Hermann
    German Canc Res Ctr, Div Clin Epidemiol & Aging Res, D-69120 Heidelberg, Germany.;German Canc Res Ctr, Natl Ctr Tumor Dis NCT, Div Prevent Oncol, D-69120 Heidelberg, Germany.;German Canc Res Ctr, German Canc Consortium DKTK, D-69120 Heidelberg, Germany..
    Brucker, Sara Y.
    Univ Tubingen, Dept Gynecol & Obstet, D-72076 Tubingen, Germany..
    Buys, Saundra S.
    Huntsman Canc Inst, Dept Med, Salt Lake City, UT 84112 USA..
    Castelao, Jose E.
    Inst Invest Sanitaria Galicia Sur IISGS, Oncol & Genet Unit, Xerencia Xest Integrada Vigo SERGAS, Vigo 36312, Spain..
    Chan, Tsun L.
    Hong Kong Hereditary Breast Canc Family Registry, Hong Kong, Peoples R China.;Hong Kong Sanat & Hosp, Dept Mol Pathol, Hong Kong, Peoples R China..
    Chang-Claude, Jenny
    German Canc Res Ctr, Div Canc Epidemiol, D-69120 Heidelberg, Germany.;Univ Canc Ctr Hamburg UCCH, Univ Med Ctr Hamburg Eppendorf, Canc Epidemiol Grp, D-20246 Hamburg, Germany..
    Chanock, Stephen J.
    NCI, Div Canc Epidemiol & Genet, US Dept HHS, NIH, Bethesda, MD 20850 USA..
    Choi, Ji-Yeob
    Seoul Natl Univ, Dept Biomed Sci, Grad Sch, Seoul 03080, South Korea.;Seoul Natl Univ, Canc Res Inst, Seoul 03080, South Korea.;Seoul Natl Univ, Inst Hlth Policy & Management, Med Res Ctr, Seoul 03080, South Korea..
    Chung, Wendy K.
    Columbia Univ, Dept Pediat, New York, NY 10032 USA. Oslo Univ Hosp, Inst Canc Res, Dept Canc Genet, Radiumhosp, N-0379 Oslo, Norway..
    Colonna, Sarah, V
    Huntsman Canc Inst, Dept Med, Salt Lake City, UT 84112 USA..
    Cornelissen, Sten
    Netherlands Canc Inst, Div Mol Pathol, Antoni Van Leeuwenhoek Hosp, NL-1066 CX Amsterdam, Netherlands..
    Couch, Fergus J.
    Mayo Clin, Dept Lab Med & Pathol, Rochester, MN 55905 USA..
    Czene, Kamila
    Karolinska Inst, Dept Med Epidemiol & Biostat, S-17165 Stockholm, Sweden..
    Daly, Mary B.
    Fox Chase Canc Ctr, Dept Clin Genet, Philadelphia, PA 19111 USA..
    Devilee, Peter
    Leiden Univ Med Ctr, Dept Pathol, NL-2333 ZA Leiden, Netherlands.;Leiden Univ Med Ctr, Dept Human Genet, NL-2333 ZA Leiden, Netherlands..
    Dork, Thilo
    Hannover Med Sch, Gynaecol Res Unit, D-30625 Hannover, Germany..
    Dossus, Laure
    Int Agcy Res Canc IARC WHO, Nutr & Metab Sect, F-69372 Lyon, France..
    Dwek, Miriam
    Univ Westminster, Sch Life Sci, London W1W 6UW, England..
    Eccles, Diana M.
    Univ Southampton, Fac Med, Southampton SO17 1BJ, Hants, England..
    Ekici, Arif B.
    Friedrich Alexander Univ Erlangen Nuremberg FAU, Univ Hosp Erlangen, Comprehens Canc Ctr Erlangen EMN, Inst Human Genet, D-91054 Erlangen, Germany..
    Eliassen, A. Heather
    Harvard Med Sch, Brigham & Womens Hosp, Dept Med, Channing Div Network Med, Boston, MA 02115 USA.;Harvard TH Chan Sch Publ Hlth, Dept Epidemiol, Boston, MA 02115 USA.;Harvard TH Chan Sch Publ Hlth, Dept Nutr, Boston, MA 02115 USA..
    Engel, Christoph
    Univ Leipzig, Inst Med Informat Stat & Epidemiol, D-04107 Leipzig, Germany.;Univ Leipzig, LIFE Leipzig Res Ctr Civilizat Dis, D-04103 Leipzig, Germany..
    Evans, D. Gareth
    Univ Manchester, Manchester Acad Hlth Sci Ctr, Fac Biol, Sch Biol Sci, Manchester, Lancs, England.;Manchester Univ NHS Fdn Trust, Manchester Acad Hlth Sci Ctr, Manchester Ctr Genom Med, North West Genom Lab Hub,St Marys Hosp, Manchester, Lancs, England..
    Fasching, Peter A.
    Friedrich Alexander Univ Erlangen Nuremberg FAU, Univ Hosp Erlangen, Comprehens Canc Ctr Erlangen EMN, Dept Gynecol & Obstet, D-91054 Erlangen, Germany.;Univ Calif Los Angeles, David Geffen Sch Med, Dept Med, Div Hematol & Oncol, Los Angeles, CA 90095 USA..
    Fletcher, Olivia
    Breast Canc Now Toby Robins Res Ctr, Inst Canc Res, London SW7 3RP, England..
    Flyger, Henrik
    Copenhagen Univ Hosp, Herlev & Gentofte Hosp, Dept Breast Surg, DK-2730 Herlev, Denmark..
    Gago-Dominguez, Manuela
    Complejo Hosp Univ Santiago, Inst Invest Sanitaria Santiago Compostela IDIS, SERGAS, Fdn Poebl Galega Med Xenom, Santiago De Compostela 15706, Spain.;Univ Calif San Diego, Moores Canc Ctr, La Jolla, CA 92037 USA..
    Gao, Yu-Tang
    Shanghai Canc Inst, Dept Epidemiol, Shanghai 20032, Peoples R China..
    Garcia-Closas, Montserrat
    NCI, Div Canc Epidemiol & Genet, US Dept HHS, NIH, Bethesda, MD 20850 USA..
    Garcia-Saenz, Jose A.
    Hosp Clin San Carlos, Ctr Invest Biomed Red Canc CIBERONC, Inst Invest Sanitaria San Carlos IdISSC, Med Oncol Dept, Madrid 28040, Spain..
    Genkinger, Jeanine
    Columbia Univ, Mailman Sch Publ Hlth, Dept Epidemiol, New York, NY 10032 USA..
    Gentry-Maharaj, Aleksandra
    UCL, Inst Clin Trials & Methodol, London WC1V 6LJ, England..
    Grassmann, Felix
    Karolinska Inst, Dept Med Epidemiol & Biostat, S-17165 Stockholm, Sweden.;Hlth & Med Univ, D-14471 Potsdam, Germany..
    Guenel, Pascal
    Univ Paris Saclay, Ctr Res Epidemiol & Populat Hlth CESP, INSERM, Team Exposome & Hered, F-94805 Villejuif, France..
    Gundert, Melanie
    German Canc Res Ctr, Mol Epidemiol Grp, C08069120, Heidelberg, Germany.;Heidelberg Univ, Univ Womens Clin Heidelberg, Mol Biol Breast Canc, D-69120 Heidelberg, Germany.;Helmholtz Zentrum Munchen, Inst Diabet Res, German Res Ctr Environm Hlth, D-85764 Neuherberg, Germany..
    Haeberle, Lothar
    Friedrich Alexander Univ Erlangen Nuremberg FAU, Univ Hosp Erlangen, Comprehens Canc Ctr Erlangen EMN, Dept Gynecol & Obstet, D-91054 Erlangen, Germany..
    Hahnen, Eric
    Univ Cologne, Univ Hosp Cologne, Fac Med, Ctr Familial Breast & Ovarian Canc, D-50937 Cologne, Germany.;Univ Cologne, Univ Hosp Cologne, Fac Med, Ctr Integrated Oncol CIO, D-50937 Cologne, Germany..
    Haiman, Christopher A.
    Univ Southern Calif, Keck Sch Med, Dept Prevent Med, Los Angeles, CA 90033 USA..
    Hakansson, Niclas
    Karolinska Inst, Inst Environm Med, S-17177 Stockholm, Sweden..
    Hall, Per
    Karolinska Inst, Dept Med Epidemiol & Biostat, S-17165 Stockholm, Sweden.;Dept Oncol, S-11883 Ssdersjukhuset, Sweden..
    Harkness, Elaine F.
    Univ Manchester, Manchester Acad Hlth Sci Ctr, Fac Biol, Div Informat, Manchester, Lancs, England.;Manchester Univ NHS Fdn Trust, Wythenshawe Hosp, Nightingale & Genesis Prevent Ctr, Manchester, Lancs, England.;Manchester Univ NHS Fdn Trust, Manchester Acad Hlth Sci Ctr, NIHR Manchester Biomed Res Unit, Manchester, Lancs, England..
    Harrington, Patricia A.
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Oncol, Cambridge, England..
    Hartikainen, Jaana M.
    Univ Eastern Finland, Translat Canc Res Area, Kuopio 70210, Finland.;Univ Eastern Finland, Inst Clin Med Pathol & Forens Med, Kuopio 70210, Finland..
    Hartman, Mikael
    Natl Univ Singapore, Natl Univ Hlth Syst, Saw Swee Hock Sch Publ Hlth, Singapore 119077, Singapore.;Natl Univ Hlth Syst, Dept Surg, Singapore 119228, Singapore..
    Hein, Alexander
    Friedrich Alexander Univ Erlangen Nuremberg FAU, Univ Hosp Erlangen, Comprehens Canc Ctr Erlangen EMN, Dept Gynecol & Obstet, D-91054 Erlangen, Germany..
    Ho, Weang-Kee
    Univ Nottingham, Dept Math Sci, Fac Sci & Engn, Malaysia Campus, Semenyih 43500, Selangor, Malaysia.;Canc Res Malaysia, Breast Canc Res Programme, Subang Jaya 47500, Selangor, Malaysia..
    Hooning, Maartje J.
    Erasmus MC Canc Inst, Dept Med Oncol, NL-3015 GD Rotterdam, Netherlands..
    Hoppe, Reiner
    Dr Margarete Fischer Bosch Inst Clin Pharmacol, D-70376 Stuttgart, Germany.;Univ Tubingen, D-72074 Tubingen, Germany..
    Hopper, John L.
    Univ Melbourne, Ctr Epidemiol & Biostat, Melbourne Sch Populat & Global Hlth, Melbourne, Vic 3010, Australia..
    Houlston, Richard S.
    Inst Canc Res, Div Genet & Epidemiol Ogy, London SM2 5NG, England..
    Howell, Anthony
    Univ Manchester, Div Canc Sci, Manchester M13 9PL, Lancs, England..
    Hunter, David J.
    Harvard TH Chan Sch Publ Hlth, Dept Epidemiol, Boston, MA 02115 USA.;Univ Oxford, Nuffield Dept Populat Hlth, Oxford OX3 7LF, England..
    Huo, Dezheng
    Univ Chicago, Ctr Clin Canc Genet, Chicago, IL 60637 USA..
    Investigators, Abctb
    Univ Sydney, Westmead Inst Med Res, Australian Breast Canc Tissue Bank, Sydney, NSW 2145, Australia..
    Ito, Hidemi
    Aichi Canc Ctr Res Inst, Div Canc Informat & Control, Nagoya, Aichi 4648681, Japan.;Nagoya Univ, Div Canc Epidemiol, Grad Sch Med, Nagoya, Aichi 4668550, Japan..
    Iwasaki, Motoki
    Natl Canc Ctr Inst Canc Control, Ctr Publ Hlth Sci, Div Epidemiol, Tokyo 1040045, Japan..
    Jakubowska, Anna
    Pomeranian Med Univ, Dept Genet & Pathol, PL-71252 Szczecin, Poland.;Pomeranian Med Univ, Independent Lab Mol Biol & Genet Diagnost, PL-71252 Szczecin, Poland..
    Janni, Wolfgang
    Univ Hosp Ulm, Dept Gynaecol & Obstet, D-89075 Ulm, Germany..
    John, Esther M.
    Stanford Univ, Sch Med, Dept Epidemiol & Populat Hlth, Stanford, CA 94305 USA.;Stanford Univ, Sch Med, Stanford Canc Inst, Dept Med, Stanford, CA 94304 USA..
    Jones, Michael E.
    Inst Canc Res, Div Genet & Epidemiol Ogy, London SM2 5NG, England..
    Jung, Audrey
    German Canc Res Ctr, Div Canc Epidemiol, D-69120 Heidelberg, Germany..
    Kaaks, Rudolf
    German Canc Res Ctr, Div Canc Epidemiol, D-69120 Heidelberg, Germany..
    Kang, Daehee
    Seoul Natl Univ, Canc Res Inst, Seoul 03080, South Korea.;Seoul Natl Univ, Coll Med, Dept Prevent Med, Seoul 03080, South Korea..
    Khusnutdinova, Elza K.
    Russian Acad Sci, Inst Biochem & Genet, Ufa Fed Res Ctr, Ufa 450054, Russia.;Bashkir State Univ, Dept Genet & Fundamental Med, Ufa 450000, Russia..
    Kim, Sung-Won
    Daerim St Marys Hosp, Dept Surg, Seoul 07442, South Korea..
    Kitahara, Cari M.
    NCI, Radiat Epidemiol Branch, Div Canc Epidemiol & Genet, Bethesda, MD 20892 USA..
    Koutros, Stella
    NCI, Div Canc Epidemiol & Genet, US Dept HHS, NIH, Bethesda, MD 20850 USA..
    Kraft, Peter
    Harvard TH Chan Sch Publ Hlth, Dept Epidemiol, Boston, MA 02115 USA.;Harvard TH Chan Sch Publ Hlth, Program Genet Epidemiol & Stat Genet, Boston, MA 02115 USA..
    Kristensen, Vessela N.
    Univ Oslo, Fac Med, Inst Clin Med, N-0450 Oslo, Norway. Vestre Viken Hosp, Dept Res, N-3019 Drammen, Norway. Oslo Univ Hosp Ullev, Dept Canc, Div Surg, Sect Breast & Endocrine Surg, N-0450 Oslo, Norway. Oslo Univ Hosp, Dept Radiol & Nucl Med, N-0379 Oslo, Norway. Akershus Univ Hosp, Dept Pathol, N-1478 Lorenskog, Norway. Oslo Univ Hosp, Inst Canc Res, Dept Tumor Biol, N-0379 Oslo, Norway. Oslo Univ Hosp, Dept Oncol, Div Surg Canc & Transplantat Med, Radiumhosp, N-0379 Oslo, Norway. Oslo Univ Hosp, Natl Advisory Unit Late Effects Canc Treatment, N-0379 Oslo, Norway. Akershus Univ Hosp, Dept Oncol, N-1478 Lorenskog, Norway. Oslo Univ Hosp, Oslo Breast Canc Res Consortium, N-0379 Oslo, Norway.;Oslo Univ Hosp, Dept Med Genet, N-0379 Oslo, Norway.;Univ Oslo, N-0379 Oslo, Norway..
    Kubelka-Sabit, Katerina
    Clin Hosp Acibadem Sistina, Dept Histopathol & Cytol, Skopje 1000, North Macedonia..
    Kurian, Allison W.
    Stanford Univ, Sch Med, Dept Epidemiol & Populat Hlth, Stanford, CA 94305 USA.;Stanford Univ, Sch Med, Stanford Canc Inst, Dept Med, Stanford, CA 94304 USA..
    Kwong, Ava
    Hong Kong Hereditary Breast Canc Family Registry, Hong Kong, Peoples R China.;Univ Hong Kong, Dept Surg, Hong Kong, Peoples R China.;Hong Kong Sanat & Hosp, Dept Surg, Hong Kong, Peoples R China.;Hong Kong Sanat & Hosp, Canc Genet Ctr, Hong Kong, Peoples R China..
    Lacey, James, V
    City Hope Natl Med Ctr, Depart Ment Computat & Quantitat Med, Duarte, CA 91010 USA.;City Hope Natl Med Ctr, City Hope Comprehens Canc Ctr, Duarte, CA 91010 USA..
    Lambrechts, Diether
    VIB Ctr Canc Biol, B-3001 Louvain, Belgium.;Univ Leuven, Dept Human Genet, Lab Translat Genet, B-3000 Louvain, Belgium..
    Le Marchand, Loic
    Univ Hawaii Canc Ctr, Epidemiol Program, Honolulu, HI 96813 USA..
    Li, Jingmei
    Genome Inst Singapore, Human Genet Div, Singapore 138672, Singapore..
    Linet, Martha
    NCI, Radiat Epidemiol Branch, Div Canc Epidemiol & Genet, Bethesda, MD 20892 USA..
    Lo, Wing-Yee
    Dr Margarete Fischer Bosch Inst Clin Pharmacol, D-70376 Stuttgart, Germany.;Univ Tubingen, D-72074 Tubingen, Germany..
    Long, Jirong
    Vanderbilt Univ, Vanderbilt Epidemiol Ctr, Vanderbilt Ingram Canc Ctr, Dept Med,Sch Med, Nashville, TN 37232 USA..
    Lophatananon, Artitaya
    Univ Manchester, Fac Biol, Sch Hlth Sci, Div Populat Hlth Hlth Serv Res & Primary Care, Manchester M13 9PL, Lancs, England..
    Mannermaa, Arto
    Univ Eastern Finland, Translat Canc Res Area, Kuopio 70210, Finland.;Univ Eastern Finland, Inst Clin Med Pathol & Forens Med, Kuopio 70210, Finland.;Kuopio Univ Hosp, Biobank Eastern Finland, Kuopio, Finland..
    Manoochehri, Mehdi
    German Canc Res Ctr, Mol Genet Breast Canc, D-69120 Heidelberg, Germany..
    Margolin, Sara
    Dept Oncol, S-11883 Ssdersjukhuset, Sweden.;Karolinska Inst, Dept Clin Sci & Educ, Sodersjukhuset, S-11883 Stockholm, Sweden..
    Matsuo, Keitaro
    Nagoya Univ, Div Canc Epidemiol, Grad Sch Med, Nagoya, Aichi 4668550, Japan.;Aichi Canc Ctr Res Inst, Div Canc Epidemiol & Prevent, Nagoya, Aichi 4648681, Japan..
    Mavroudis, Dimitrios
    Univ Hosp Heraklion, Dept Med Oncol, Iraklion 71110, Greece..
    Menon, Usha
    UCL, Inst Clin Trials & Methodol, London WC1V 6LJ, England..
    Muir, Kenneth
    Univ Manchester, Fac Biol, Sch Hlth Sci, Div Populat Hlth Hlth Serv Res & Primary Care, Manchester M13 9PL, Lancs, England..
    Murphy, Rachel A.
    Univ British Columbia, Sch Populat & Publ Hlth, Vancouver, BC V6T 1Z4, Canada.;BC Canc, Canc Control Res, Vancouver, BC V5Z 1L3, Canada..
    Nevanlinna, Heli
    Univ Helsinki, Helsinki Univ Hosp, Dept Obstet & Gynecol, Helsinki 00290, Finland..
    Newman, William G.
    Univ Manchester, Manchester Acad Hlth Sci Ctr, Fac Biol, Sch Biol Sci, Manchester, Lancs, England.;Manchester Univ NHS Fdn Trust, Manchester Acad Hlth Sci Ctr, Manchester Ctr Genom Med, North West Genom Lab Hub,St Marys Hosp, Manchester, Lancs, England..
    Niederacher, Dieter
    Heinrich Heine Univ Dusseldorf, Univ Hosp Dusseldorf, Dept Gynecol & Obstet, D-40225 Dusseldorf, Germany..
    O'Brien, Katie M.
    NIEHS, Epidemiol Branch, NIH, Res Triangle Pk, NC 27709 USA..
    Obi, Nadia
    Univ Med Ctr Hamburg Eppendorf, Inst Med Biometry & Epidemiol, D-20246 Hamburg, Germany..
    Offit, Kenneth
    Mem Sloan Kettering Canc Ctr, Dept Canc Biol & Genet, Clin Genet Res Lab, New York, NY 10065 USA.;Mem Sloan Kettering Canc Ctr, Dept Med, Clin Genet Serv, New York, NY 10065 USA..
    Olopade, Olufunmilayo, I
    Univ Chicago, Ctr Clin Canc Genet, Chicago, IL 60637 USA..
    Olshan, Andrew F.
    Univ North Carolina Chapel Hill, Dept Epidemiol, Gillings Sch Global Publ Hlth, Chapel Hill, NC USA.;Univ North Carolina Chapel Hill, UNC Lineberger Comprehens Canc Ctr, Chapel Hill, NC USA..
    Olsson, Hakan
    Lund Univ, Dept Canc Epidemiol, Clin Sci, S-22242 Lund, Sweden..
    Park, Sue K.
    Seoul Natl Univ, Canc Res Inst, Seoul 03080, South Korea.;Seoul Natl Univ, Coll Med, Dept Prevent Med, Seoul 03080, South Korea.;Seoul Natl Univ, Coll Med, Integrated Major Innovat Med Sci, Seoul 03080, South Korea..
    Patel, Alpa, V
    Amer Canc Soc, Dept Populat Sci, Atlanta, GA 30303 USA..
    Patel, Achal
    Univ North Carolina Chapel Hill, Dept Epidemiol, Gillings Sch Global Publ Hlth, Chapel Hill, NC USA.;Univ North Carolina Chapel Hill, UNC Lineberger Comprehens Canc Ctr, Chapel Hill, NC USA..
    Perou, Charles M.
    Univ North Carolina Chapel Hill, Dept Genet, Lineberger Comprehens Canc Ctr, Chapel Hill, NC USA..
    Peto, Julian
    London Sch Hyg & Trop Med, Dept Noncommunicable Dis Epidemiol, London WC1E 7HT, England..
    Pharoah, Paul D. P.
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Publ Hlth & Primary Care, Cambridge CB1 8RN, England.;Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Oncol, Cambridge, England..
    Plaseska-Karanfilska, Dijana
    MASA, Res Ctr Genet Engn & Biotechnol Georgi D Efremov, Skopje 1000, North Macedonia..
    Presneau, Nadege
    Univ Westminster, Sch Life Sci, London W1W 6UW, England..
    Rack, Brigitte
    Univ Hosp Ulm, Dept Gynaecol & Obstet, D-89075 Ulm, Germany..
    Radice, Paolo
    Fdn IRCCS Ist Nazl Tumori INT, Dept Res, Unit Mol Bases Genet Risk & Genet Testing, I-20133 Milan, Italy..
    Ramachandran, Dhanya
    Hannover Med Sch, Gynaecol Res Unit, D-30625 Hannover, Germany..
    Rashid, Muhammad U.
    German Canc Res Ctr, Mol Genet Breast Canc, D-69120 Heidelberg, Germany.;Shaukat Khanum Mem Canc Hosp & Res Ctr SKMCH & RC, Dept Basic Sci, Lahore 54000, Pakistan..
    Rennert, Gad
    Carmel Hosp, Clalit Natl Canc Control Ctr, IL-35254 Haifa, Israel.;Technion Fac Med, IL-35254 Haifa, Israel..
    Romero, Atocha
    Hosp Univ Puerta de Hierro, Med Oncol Dept, Madrid 28222, Spain..
    Ruddy, Kathryn J.
    Mayo Clin, Dept Oncol, Rochester, MN 55905 USA..
    Ruebner, Matthias
    Friedrich Alexander Univ Erlangen Nuremberg FAU, Univ Hosp Erlangen, Comprehens Canc Ctr Erlangen EMN, Dept Gynecol & Obstet, D-91054 Erlangen, Germany..
    Saloustros, Emmanouil
    Univ Hosp Larissa, Dept Oncol, Larisa 41110, Greece..
    Sandler, Dale P.
    NIEHS, Epidemiol Branch, NIH, Res Triangle Pk, NC 27709 USA..
    Sawyer, Elinor J.
    Kings Coll London, Comprehens Canc Ctr, Sch Canc & Pharmaceut Sci, Guys Campus, London, England..
    Schmidt, Marjanka K.
    Netherlands Canc Inst, Div Mol Pathol, Antoni Van Leeuwenhoek Hosp, NL-1066 CX Amsterdam, Netherlands.;Antoni Van Leeuwenhoek Hosp, Div Psychosocial Res & Epidemiol, Netherlands Canc Inst, NL-1066 CX Amsterdam, Netherlands..
    Schmutzler, Rita K.
    Univ Cologne, Univ Hosp Cologne, Fac Med, Ctr Familial Breast & Ovarian Canc, D-50937 Cologne, Germany.;Univ Cologne, Univ Hosp Cologne, Fac Med, Ctr Integrated Oncol CIO, D-50937 Cologne, Germany.;Univ Cologne, Fac Med, Ctr Mol Med Cologne CMMC, D-50931 Cologne, Germany.;Univ Cologne, Univ Hosp Cologne, D-50931 Cologne, Germany..
    Schneider, Michael O.
    Friedrich Alexander Univ Erlangen Nuremberg FAU, Univ Hosp Erlangen, Comprehens Canc Ctr Erlangen EMN, Dept Gynecol & Obstet, D-91054 Erlangen, Germany..
    Scott, Christopher
    Mayo Clin, Dept Hlth Sci Res, Rochester, MN 55905 USA..
    Shah, Mitul
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Oncol, Cambridge, England..
    Sharma, Priyanka
    Univ Kansas Med Ctr, Dept Internal Med, Div Med Oncol, Westwood, KS 66205 USA..
    Shen, Chen-Yang
    Acad Sinica, Inst Biomed Sci, Taipei 115, Taiwan.;China Med Univ, Sch Publ Hlth, Taichung, Taiwan..
    Shu, Xiao-Ou
    Vanderbilt Univ, Vanderbilt Epidemiol Ctr, Vanderbilt Ingram Canc Ctr, Dept Med,Sch Med, Nashville, TN 37232 USA..
    Simard, Jacques
    Ctr Hosp Univ Quebec Univ Laval Res Ctr, Genom Ctr, Quebec City, PQ G1V 4G2, Canada..
    Surowy, Harald
    German Canc Res Ctr, Mol Epidemiol Grp, C08069120, Heidelberg, Germany.;Heidelberg Univ, Univ Womens Clin Heidelberg, Mol Biol Breast Canc, D-69120 Heidelberg, Germany..
    Tamimi, Rulla M.
    Harvard TH Chan Sch Publ Hlth, Dept Epidemiol, Boston, MA 02115 USA.;Weill Cornell Med, Dept Populat Hlth Sci, New York, NY 10065 USA..
    Tapper, William J.
    Univ Southampton, Fac Med, Southampton SO17 1BJ, Hants, England..
    Taylor, Jack A.
    NIEHS, Epidemiol Branch, NIH, Res Triangle Pk, NC 27709 USA.;NIEHS, Epigenet & Stem Cell Biol Lab, NIH, Res Triangle Pk, NC 27709 USA..
    Teo, Soo Hwang
    Canc Res Malaysia, Breast Canc Res Programme, Subang Jaya 47500, Selangor, Malaysia.;Univ Malaya, Fac Med, Dept Surg, Kuala Lumpur 50603, Malaysia..
    Teras, Lauren R.
    Amer Canc Soc, Dept Populat Sci, Atlanta, GA 30303 USA..
    Toland, Amanda E.
    Ohio State Univ, Dept Canc Biol & Genet, Columbus, OH 43210 USA..
    Tollenaar, Rob A. E. M.
    Leiden Univ Med Ctr, Dept Surg, NL-2333 ZA Leiden, Netherlands..
    Torres, Diana
    German Canc Res Ctr, Mol Genet Breast Canc, D-69120 Heidelberg, Germany.;Pontificia Univ Javeriana, Inst Human Genet, Bogota 110231, Colombia..
    Torres-Mejia, Gabriela
    Ctr Populat Hlth Res, Natl Inst Publ Hlth, Cuernavaca 62100, Morelos, Mexico..
    Troester, Melissa A.
    Univ North Carolina Chapel Hill, Dept Epidemiol, Gillings Sch Global Publ Hlth, Chapel Hill, NC USA.;Univ North Carolina Chapel Hill, UNC Lineberger Comprehens Canc Ctr, Chapel Hill, NC USA..
    Truong, Therese
    Univ Paris Saclay, Ctr Res Epidemiol & Populat Hlth CESP, INSERM, Team Exposome & Hered, F-94805 Villejuif, France..
    Vachon, Celine M.
    Mayo Clin, Div Epidemiol, Dept Quantitat Hlth Sci, Rochester, MN 55905 USA..
    Vijai, Joseph
    Mem Sloan Kettering Canc Ctr, Dept Canc Biol & Genet, Clin Genet Res Lab, New York, NY 10065 USA.;Mem Sloan Kettering Canc Ctr, Dept Med, Clin Genet Serv, New York, NY 10065 USA..
    Weinberg, Clarice R.
    NIEHS, Biostat & Computat Biol Branch, NIH, Res Triangle Pk, NC 27709 USA..
    Wendt, Camilla
    Karolinska Inst, Dept Clin Sci & Educ, Sodersjukhuset, S-11883 Stockholm, Sweden..
    Winqvist, Robert
    Univ Oulu, Bioctr Oulu, Canc & Translat Med Res Unit, Lab Canc Genet & Tumor Biol, Oulu 90570, Finland.;Northern Finland Lab Ctr Oulu, Lab Canc Genet & Tumor Biol, Oulu 90570, Finland..
    Wolk, Alicja
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Surgical Sciences. Karolinska Inst, Inst Environm Med, S-17177 Stockholm, Sweden..
    Wu, Anna H.
    Univ Southern Calif, Keck Sch Med, Dept Prevent Med, Los Angeles, CA 90033 USA..
    Yamaji, Taiki
    Natl Canc Ctr Inst Canc Control, Ctr Publ Hlth Sci, Div Epidemiol, Tokyo 1040045, Japan..
    Yang, Xiaohong R.
    NCI, Div Canc Epidemiol & Genet, US Dept HHS, NIH, Bethesda, MD 20850 USA..
    Yu, Jyh-Cherng
    Triserv Gen Hosp, Natl Def Med Ctr, Dept Surg, Taipei 114, Taiwan..
    Zheng, Wei
    Vanderbilt Univ, Vanderbilt Epidemiol Ctr, Vanderbilt Ingram Canc Ctr, Dept Med,Sch Med, Nashville, TN 37232 USA..
    Ziogas, Argyrios
    Univ Calif Irvine, Genet Epidemiol Res Inst, Dept Med, Irvine, CA 92617 USA..
    Ziv, Elad
    Univ Calif San Francisco, Diller Family Comprehens Canc Ctr, Inst Human Genet, Dept Med, San Francisco, CA 94115 USA..
    Dunning, Alison M.
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Oncol, Cambridge, England..
    Easton, Douglas F.
    Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Publ Hlth & Primary Care, Cambridge CB1 8RN, England.;Univ Cambridge, Ctr Canc Genet Epidemiol, Dept Oncol, Cambridge, England..
    Hemingway, Harry
    UCL, Inst Hlth Informat, London, England.;UCL, Hlth Data Res UK, London, England.;Univ Coll London Hosp, Biomed Res Ctr UCLH BRC, London, England.;Alan Turing Inst, London, England..
    Hamann, Ute
    German Canc Res Ctr, Mol Genet Breast Canc, D-69120 Heidelberg, Germany..
    Kuchenbaecker, Karoline B.
    UCL, Div Psychiat, London, England.;UCL, UCL Genet Inst, London, England..
    Aggregation tests identify new gene associations with breast cancer in populations with diverse ancestry2023In: Genome Medicine, E-ISSN 1756-994X, Vol. 15, article id 7Article in journal (Refereed)
    Abstract [en]

    Background: Low-frequency variants play an important role in breast cancer (BC) susceptibility. Gene-based methods can increase power by combining multiple variants in the same gene and help identify target genes.

    Methods: We evaluated the potential of gene-based aggregation in the Breast Cancer Association Consortium cohorts including 83,471 cases and 59,199 controls. Low-frequency variants were aggregated for individual genes' coding and regulatory regions. Association results in European ancestry samples were compared to single-marker association results in the same cohort. Gene-based associations were also combined in meta-analysis across individuals with European, Asian, African, and Latin American and Hispanic ancestry.

    Results: In European ancestry samples, 14 genes were significantly associated (q < 0.05) with BC. Of those, two genes, FMNL3 (P = 6.11 x 10(-6)) and AC058822.1 (P = 1.47 x 10(-4)), represent new associations. High FMNL3 expression has previously been linked to poor prognosis in several other cancers. Meta-analysis of samples with diverse ancestry discovered further associations including established candidate genes ESR1 and CBLB. Furthermore, literature review and database query found further support for a biologically plausible link with cancer for genes CBLB, FMNL3, FGFR2, LSP1, MAP3K1, and SRGAP2C.

    Conclusions: Using extended gene-based aggregation tests including coding and regulatory variation, we report identification of plausible target genes for previously identified single-marker associations with BC as well as the discovery of novel genes implicated in BC development. Including multi ancestral cohorts in this study enabled the identification of otherwise missed disease associations as ESR1 (P = 1.31 x 10(-5)), demonstrating the importance of diversifying study cohorts.

    Download full text (pdf)
    FULLTEXT01
  • 32.
    Oresic, Matej
    et al.
    Örebro University, School of Medical Sciences. VTT Technical Research Centre of Finland, Espoo, Finland.
    Lötjönen, Jyrki
    VTT Technical Research Centre of Finland, Tampere, Finland.
    Soininen, Hilkka
    Kuopio University Hospital, University of Eastern Finland, Kuopio, Finland.
    Systems medicine and the integration of bioinformatic tools for the diagnosis of Alzheimer's disease2010In: Genome Medicine, E-ISSN 1756-994X, Vol. 2, no 11, article id 83Article in journal (Refereed)
    Abstract [en]

    Because of the changes in demographic structure, the prevalence of Alzheimer's disease is expected to rise dramatically over the next decades. The progression of this degenerative and terminal disease is gradual, with the subclinical stage of illness believed to span several decades. Despite this, no therapy to prevent or cure Alzheimer's disease is currently available. Early disease detection is still important for delaying the onset of the disease with pharmacological treatment and/or lifestyle changes, assessing the efficacy of potential therapeutic agents, or monitoring disease progression more closely using medical imaging. Sensitive cerebrospinal-fluid-derived marker candidates exist, but given the invasiveness of sample collection their use in routine diagnostics may be limited. The pathogenesis of Alzheimer's disease is complex and poorly understood. There is thus a strong case for integrating information across multiple physiological levels, from molecular profiling (metabolomics, lipidomics, proteomics and transcriptomics) and brain imaging to cognitive assessments. To facilitate the integration of heterogeneous data, such as molecular and image data, sophisticated statistical approaches are needed to segment the image data and study their dependencies on molecular changes in the same individuals. Molecular profiling, combined with biophysical modeling of molecular assemblies associated with the disease, offer an opportunity to link the molecular pathway changes with cell- and tissue-level physiology and structure. Given that data acquired at different levels can carry complementary information about early Alzheimer's disease pathology, it is expected that their integration will improve early detection as well as our understanding of the disease.

  • 33.
    Oresic, Matej
    et al.
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Seppänen-Laakso, T.
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Sun, D.
    Department of Psychology, University of California, Los Angeles CA, United States.
    Tang, J.
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Therman, S.
    Department of Mental Health and Alcohol Research, National Institute for Health and Welfare, Helsinki, Finland.
    Viehman, R.
    Department of Psychology, University of California, Los Angeles CA, United States.
    Mustonen, U.
    Department of Mental Health and Alcohol Research, National Institute for Health and Welfare, Helsinki, Finland.
    van Erp, T. G.
    Department of Psychiatry and Human Behavior, University of California Irvine, Irvine CA, United States.
    Hyötyläinen, Tuulia
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Thompson, P.
    Department of Neurology and Laboratory of NeuroImaging, University of California, Los Angeles CA, United States.
    Toga, A. W.
    Department of Neurology and Laboratory of NeuroImaging, University of California, Los Angeles CA, United States.
    Huttunen, M. O.
    Department of Mental Health and Alcohol Research, National Institute for Health and Welfare, Helsinki, Finland.
    Suvisaari, J.
    Department of Mental Health and Alcohol Research, National Institute for Health and Welfare, Helsinki, Finland.
    Kaprio, J.
    Department of Mental Health and Alcohol Research, National Institute for Health and Welfare, Helsinki, Finland; Department of Public Health, University of Helsinki, Helsinki, Finland.
    Lönnqvist, J.
    Department of Mental Health and Alcohol Research, National Institute for Health and Welfare, Helsinki, Finland; Department of Psychiatry, Helsinki University Hospital, Helsinki, Finland.
    Cannon, T. D.
    Department of Psychology, University of California, Los Angeles CA, United States; Department of Psychiatry and Biobehavioral Sciences, University of California Los, Los Angeles CA, United States.
    Phospholipids and insulin resistance in psychosis: A lipidomics study of twin pairs discordant for schizophrenia2012In: Genome Medicine, E-ISSN 1756-994X, Vol. 4, no 1, article id 1Article in journal (Refereed)
    Abstract [en]

    Background: Several theories have been proposed to conceptualize the pathological processes inherent to schizophrenia. The 'prostaglandin deficiency' hypothesis postulates that defective enzyme systems converting essential fatty acids to prostaglandins lead to diminished levels of prostaglandins, which in turn affect synaptic transmission.

    Methods: Here we sought to determine the lipidomic profiles associated with schizophrenia in twin pairs discordant for schizophrenia as well as unaffected twin pairs. The study included serum samples from 19 twin pairs discordant for schizophrenia (mean age 51 +/- 10 years; 7 monozygotic pairs; 13 female pairs) and 34 age and gender matched healthy twins as controls. Neurocognitive assessment data and gray matter density measurements taken from high-resolution magnetic resonance images were also obtained. A lipidomics platform using ultra performance liquid chromatography coupled to time-of-flight mass spectrometry was applied for the analysis of serum samples.

    Results: In comparison to their healthy co-twins, the patients had elevated triglycerides and were more insulin resistant. They had diminished lysophosphatidylcholine levels, which associated with decreased cognitive speed.

    Conclusions: Our findings may be of pathophysiological relevance since lysophosphatidylcholines, byproducts of phospholipase A2-catalyzed phospholipid hydrolysis, are preferred carriers of polyunsaturated fatty acids across the blood-brain barrier. Furthermore, diminishment of lysophosphatidylcholines suggests that subjects at risk of schizophrenia may be more susceptible to infections. Their association with cognitive speed supports the view that altered neurotransmission in schizophrenia may be in part mediated by reactive lipids such as prostaglandins.

  • 34.
    Oresic, Matej
    et al.
    Örebro University, School of Medical Sciences. VTT Technical Research Centre of Finland, Espoo, Finland.
    Tang, Jing
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Seppänen-Laakso, Tuulikki
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Mattila, Ismo
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Saarni, Suoma E
    National Institute for Health and Welfare, Helsinki, Finland.
    Saarni, Samuli I.
    National Institute for Health and Welfare, Helsinki, Finland; Department of Psychiatry, Helsinki University Central Hospital, Helsinki, Finland.
    Lönnqvist, Jouko
    National Institute for Health and Welfare, Helsinki, Finland; Department of Psychiatry, Helsinki University Central Hospital, Helsinki, Finland.
    Sysi-Aho, Marko
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Hyötyläinen, Tuulia
    Örebro University, School of Science and Technology. VTT Technical Research Centre of Finland, Espoo, Finland.
    Perälä, Jonna
    National Institute for Health and Welfare, Helsinki, Finland.
    Suvisaari, Jaana
    National Institute for Health and Welfare, Helsinki, Finland.
    Metabolome in schizophrenia and other psychotic disorders: a general population-based study2011In: Genome Medicine, E-ISSN 1756-994X, Vol. 3, no 3, article id 19Article in journal (Refereed)
    Abstract [en]

    BACKGROUND: Persons with schizophrenia and other psychotic disorders have a high prevalence of obesity, impaired glucose tolerance, and lipid abnormalities, particularly hypertriglyceridemia and low high-density lipoprotein. More detailed molecular information on the metabolic abnormalities may reveal clues about the pathophysiology of these changes, as well as about disease specificity.

    METHODS: We applied comprehensive metabolomics in serum samples from a general population-based study in Finland. The study included all persons with DSM-IV primary psychotic disorder (schizophrenia, n = 45; other non-affective psychosis (ONAP), n = 57; affective psychosis, n = 37) and controls matched by age, sex, and region of residence. Two analytical platforms for metabolomics were applied to all serum samples: a global lipidomics platform based on ultra-performance liquid chromatography coupled to mass spectrometry, which covers molecular lipids such as phospholipids and neutral lipids; and a platform for small polar metabolites based on two-dimensional gas chromatography coupled to time-of-flight mass spectrometry (GC × GC-TOFMS).

    RESULTS: Compared with their matched controls, persons with schizophrenia had significantly higher metabolite levels in six lipid clusters containing mainly saturated triglycerides, and in two small-molecule clusters containing, among other metabolites, (1) branched chain amino acids, phenylalanine and tyrosine, and (2) proline, glutamic, lactic and pyruvic acids. Among these, serum glutamic acid was elevated in all psychoses (P = 0.0020) compared to controls, while proline upregulation (P = 0.000023) was specific to schizophrenia. After adjusting for medication and metabolic comorbidity in linear mixed models, schizophrenia remained independently associated with higher levels in seven of these eight clusters (P < 0.05 in each cluster). The metabolic abnormalities were less pronounced in persons with ONAP or affective psychosis.

    CONCLUSIONS: Our findings suggest that specific metabolic abnormalities related to glucoregulatory processes and proline metabolism are specifically associated with schizophrenia and reflect two different disease-related pathways. Metabolomics, which is sensitive to both genetic and environmental variation, may become a powerful tool in psychiatric research to investigate disease susceptibility, clinical course, and treatment response.

  • 35.
    Schäfer, Samuel
    et al.
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center for Surgery, Orthopaedics and Cancer Treatment, Mag- tarmmedicinska kliniken.
    Smelik, Martin
    Karolinska Inst, Sweden.
    Sysoev, Oleg
    Linköping University, Department of Computer and Information Science, The Division of Statistics and Machine Learning. Linköping University, Faculty of Arts and Sciences.
    Zhao, Yelin
    Karolinska Inst, Sweden.
    Eklund, Desiré
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Neurobiology. Linköping University, Faculty of Medicine and Health Sciences.
    Lilja, Sandra
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences. Mavatar Inc, Sweden.
    Gustafsson, Mika
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Heyn, Holger
    Barcelona Inst Sci & Technol BIST, Spain; Univ Pompeu Fabra UPF, Spain.
    Julia, Antonio
    Inst Recerca Vall dHebron, Spain.
    Kovacs, Istvan A.
    Northwestern Univ, IL 60208 USA; Northwestern Univ, IL 60208 USA.
    Loscalzo, Joseph
    Brigham & Womens Hosp, MA USA.
    Marsal, Sara
    Inst Recerca Vall dHebron, Spain.
    Zhang, Huan
    Linköping University, Department of Biomedical and Clinical Sciences, Division of Children's and Women's Health. Linköping University, Faculty of Medicine and Health Sciences.
    Li, Xinxiu
    Karolinska Inst, Sweden.
    Gawel, Danuta
    Mavatar Inc, Sweden.
    Wang, Hui
    Karolinska Inst, Sweden; Xuzhou Med Univ, Peoples R China.
    Benson, Mikael
    Karolinska Inst, Sweden.
    scDrugPrio: a framework for the analysis of single-cell transcriptomics to address multiple problems in precision medicine in immune-mediated inflammatory diseases2024In: Genome Medicine, E-ISSN 1756-994X, Vol. 16, no 1, article id 42Article in journal (Refereed)
    Abstract [en]

    Background Ineffective drug treatment is a major problem for many patients with immune-mediated inflammatory diseases (IMIDs). Important reasons are the lack of systematic solutions for drug prioritisation and repurposing based on characterisation of the complex and heterogeneous cellular and molecular changes in IMIDs.Methods Here, we propose a computational framework, scDrugPrio, which constructs network models of inflammatory disease based on single-cell RNA sequencing (scRNA-seq) data. scDrugPrio constructs detailed network models of inflammatory diseases that integrate information on cell type-specific expression changes, altered cellular crosstalk and pharmacological properties for the selection and ranking of thousands of drugs.Results scDrugPrio was developed using a mouse model of antigen-induced arthritis and validated by improved precision/recall for approved drugs, as well as extensive in vitro, in vivo, and in silico studies of drugs that were predicted, but not approved, for the studied diseases. Next, scDrugPrio was applied to multiple sclerosis, Crohn's disease, and psoriatic arthritis, further supporting scDrugPrio through prioritisation of relevant and approved drugs. However, in contrast to the mouse model of arthritis, great interindividual cellular and gene expression differences were found in patients with the same diagnosis. Such differences could explain why some patients did or did not respond to treatment. This explanation was supported by the application of scDrugPrio to scRNA-seq data from eleven individual Crohn's disease patients. The analysis showed great variations in drug predictions between patients, for example, assigning a high rank to anti-TNF treatment in a responder and a low rank in a nonresponder to that treatment.Conclusions We propose a computational framework, scDrugPrio, for drug prioritisation based on scRNA-seq of IMID disease. Application to individual patients indicates scDrugPrio's potential for personalised network-based drug screening on cellulome-, genome-, and drugome-wide scales. For this purpose, we made scDrugPrio into an easy-to-use R package (https://github.com/SDTC-CPMed/scDrugPrio).

  • 36.
    Serra-Musach, Jordi
    et al.
    Bellvitge Institute Biomed Research IDIBELL, Spain.
    Mateo, Francesca
    Bellvitge Institute Biomed Research IDIBELL, Spain.
    Capdevila-Busquets, Eva
    Barcelona Institute Science and Technology, Spain.
    Ruiz de Garibay, Gorka
    Bellvitge Institute Biomed Research IDIBELL, Spain.
    Zhang, Xiaohu
    NIH, MD 20850 USA.
    Guha, Raj
    NIH, MD 20850 USA.
    Thomas, Craig J.
    NIH, MD 20850 USA.
    Grueso, Judit
    VHIO, Spain.
    Villanueva, Alberto
    Bellvitge Institute Biomed Research IDIBELL, Spain.
    Jaeger, Samira
    Barcelona Institute Science and Technology, Spain.
    Heyn, Holger
    IDIBELL, Spain.
    Vizoso, Miguel
    IDIBELL, Spain.
    Perez, Hector
    IDIBELL, Spain.
    Cordero, Alex
    IDIBELL, Spain.
    Gonzalez-Suarez, Eva
    IDIBELL, Spain.
    Esteller, Manel
    IDIBELL, Spain; University of Barcelona, Spain; University of Barcelona, Spain; ICREA, Spain.
    Moreno-Bueno, Gema
    Autonomous University of Madrid, Spain; MD Anderson Int Fdn, Spain.
    Tjärnberg, Andreas
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Lazaro, Conxi
    IDIBELL, Spain.
    Serra, Violeta
    VHIO, Spain.
    Arribas, Joaquin
    ICREA, Spain; VHIO, Spain; Autonomous University of Barcelona, Spain.
    Benson, Mikael
    Linköping University, Department of Clinical and Experimental Medicine, Division of Clinical Sciences. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Heart and Medicine Center, Allergy Center.
    Gustafsson, Mika
    Linköping University, Department of Physics, Chemistry and Biology, Bioinformatics. Linköping University, Faculty of Science & Engineering.
    Ferrer, Marc
    NIH, MD 20850 USA.
    Aloy, Patrick
    Barcelona Institute Science and Technology, Spain; ICREA, Spain.
    Angel Pujana, Miquel
    Bellvitge Institute Biomed Research IDIBELL, Spain.
    Cancer network activity associated with therapeutic response and synergism2016In: Genome Medicine, E-ISSN 1756-994X, Vol. 8, no 88Article in journal (Refereed)
    Abstract [en]

    Background: Cancer patients often show no or only modest benefit from a given therapy. This major problem in oncology is generally attributed to the lack of specific predictive biomarkers, yet a global measure of cancer cell activity may support a comprehensive mechanistic understanding of therapy efficacy. We reasoned that network analysis of omic data could help to achieve this goal. Methods: A measure of "cancer network activity" (CNA) was implemented based on a previously defined network feature of communicability. The network nodes and edges corresponded to human proteins and experimentally identified interactions, respectively. The edges were weighted proportionally to the expression of the genes encoding for the corresponding proteins and relative to the number of direct interactors. The gene expression data corresponded to the basal conditions of 595 human cancer cell lines. Therapeutic responses corresponded to the impairment of cell viability measured by the half maximal inhibitory concentration (IC50) of 130 drugs approved or under clinical development. Gene ontology, signaling pathway, and transcription factor-binding annotations were taken from public repositories. Predicted synergies were assessed by determining the viability of four breast cancer cell lines and by applying two different analytical methods. Results: The effects of drug classes were associated with CNAs formed by different cell lines. CNAs also differentiate target families and effector pathways. Proteins that occupy a central position in the network largely contribute to CNA. Known key cancer-associated biological processes, signaling pathways, and master regulators also contribute to CNA. Moreover, the major cancer drivers frequently mediate CNA and therapeutic differences. Cell-based assays centered on these differences and using uncorrelated drug effects reveals novel synergistic combinations for the treatment of breast cancer dependent on PI3K-mTOR signaling. Conclusions: Cancer therapeutic responses can be predicted on the basis of a systems-level analysis of molecular interactions and gene expression. Fundamental cancer processes, pathways, and drivers contribute to this feature, which can also be exploited to predict precise synergistic drug combinations.

    Download full text (pdf)
    fulltext
  • 37.
    Stranneheim, Henrik
    et al.
    Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Univ Hosp, Ctr Inherited Metab Dis, Stockholm, Sweden.;Karolinska Inst, Dept Microbiol Tumour & Cell Biol, Sci Life Lab, Stockholm, Sweden..
    Laaksonen, Mikael
    KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH Royal Inst Technol, Sch Engn Sci Chem Biotechnol & Hlth, Sci Life Lab, Stockholm, Sweden..
    Rosenbaum, Adam
    KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Wirta, Valtteri
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Gene Technology. Karolinska Inst, Dept Microbiol Tumour & Cell Biol, Sci Life Lab, Stockholm, Sweden..
    Wedell, Anna
    Karolinska Inst, Dept Mol Med & Surg, Stockholm, Sweden.;Karolinska Univ Hosp, Ctr Inherited Metab Dis, Stockholm, Sweden.;Karolinska Inst, Dept Mol Med & Surg, Sci Life Lab, Stockholm, Sweden..
    et al.,
    Integration of whole genome sequencing into a healthcare setting: high diagnostic rates across multiple clinical entities in 3219 rare disease patients2021In: Genome Medicine, E-ISSN 1756-994X, Vol. 13, no 1, article id 40Article in journal (Refereed)
    Abstract [en]

    Background We report the findings from 4437 individuals (3219 patients and 1218 relatives) who have been analyzed by whole genome sequencing (WGS) at the Genomic Medicine Center Karolinska-Rare Diseases (GMCK-RD) since mid-2015. GMCK-RD represents a long-term collaborative initiative between Karolinska University Hospital and Science for Life Laboratory to establish advanced, genomics-based diagnostics in the Stockholm healthcare setting. Methods Our analysis covers detection and interpretation of SNVs, INDELs, uniparental disomy, CNVs, balanced structural variants, and short tandem repeat expansions. Visualization of results for clinical interpretation is carried out in Scout-a custom-developed decision support system. Results from both singleton (84%) and trio/family (16%) analyses are reported. Variant interpretation is done by 15 expert teams at the hospital involving staff from three clinics. For patients with complex phenotypes, data is shared between the teams. Results Overall, 40% of the patients received a molecular diagnosis ranging from 19 to 54% for specific disease groups. There was heterogeneity regarding causative genes (n = 754) with some of the most common ones being COL2A1 (n = 12; skeletal dysplasia), SCN1A (n = 8; epilepsy), and TNFRSF13B (n = 4; inborn errors of immunity). Some causative variants were recurrent, including previously known founder mutations, some novel mutations, and recurrent de novo mutations. Overall, GMCK-RD has resulted in a large number of patients receiving specific molecular diagnoses. Furthermore, negative cases have been included in research studies that have resulted in the discovery of 17 published, novel disease-causing genes. To facilitate the discovery of new disease genes, GMCK-RD has joined international data sharing initiatives, including ClinVar, UDNI, Beacon, and MatchMaker Exchange. Conclusions Clinical WGS at GMCK-RD has provided molecular diagnoses to over 1200 individuals with a broad range of rare diseases. Consolidation and spread of this clinical-academic partnership will enable large-scale national collaboration.

  • 38.
    Sun, Jing
    et al.
    Zhejiang Univ, Dept Big Data Hlth Sci, Sch Publ Hlth, Sch Med, Hangzhou, Zhejiang, Peoples R China.;Zhejiang Univ, Analyt Affiliated Hosp 2, Ctr Clin Big Data, Sch Med, Hangzhou, Zhejiang, Peoples R China..
    Zhao, Jianhui
    Zhejiang Univ, Dept Big Data Hlth Sci, Sch Publ Hlth, Sch Med, Hangzhou, Zhejiang, Peoples R China.;Zhejiang Univ, Analyt Affiliated Hosp 2, Ctr Clin Big Data, Sch Med, Hangzhou, Zhejiang, Peoples R China..
    Jiang, Fangyuan
    Zhejiang Univ, Dept Big Data Hlth Sci, Sch Publ Hlth, Sch Med, Hangzhou, Zhejiang, Peoples R China.;Zhejiang Univ, Analyt Affiliated Hosp 2, Ctr Clin Big Data, Sch Med, Hangzhou, Zhejiang, Peoples R China..
    Wang, Lijuan
    Univ Edinburgh, Usher Inst, Ctr Global Hlth, Edinburgh, Scotland..
    Xiao, Qian
    Zhejiang Univ, Affiliated Hosp 2, Colorectal Surg & Oncol, Key Lab Canc Prevent & Intervent,Minist Educ,Sch M, Hangzhou, Peoples R China..
    Han, Fengyan
    Zhejiang Univ, Sch Med, Womens Hosp, Dept Pathol, Hangzhou, Zhejiang, Peoples R China..
    Chen, Jie
    Zhejiang Univ, Dept Big Data Hlth Sci, Sch Publ Hlth, Sch Med, Hangzhou, Zhejiang, Peoples R China.;Zhejiang Univ, Analyt Affiliated Hosp 2, Ctr Clin Big Data, Sch Med, Hangzhou, Zhejiang, Peoples R China..
    Yuan, Shuai
    Karolinska Inst, Inst Environm Med, Unit Cardiovasc & Nutr Epidemiol, Stockholm, Sweden..
    Wei, Jingsun
    Zhejiang Univ, Affiliated Hosp 2, Colorectal Surg & Oncol, Key Lab Canc Prevent & Intervent,Minist Educ,Sch M, Hangzhou, Peoples R China..
    Larsson, Susanna C.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Surgical Sciences, Medical epidemiology. Karolinska Inst, Inst Environm Med, Unit Cardiovasc & Nutr Epidemiol, Stockholm, Sweden..
    Zhang, Honghe
    Zhejiang Univ, Sch Med, Womens Hosp, Dept Pathol, Hangzhou, Zhejiang, Peoples R China..
    Dunlop, Malcolm G.
    Univ Edinburgh, Med Res Council Inst Genet & Canc, Canc Res UK Edinburgh Ctr, Edinburgh, Scotland.;Univ Edinburgh, Inst Genet & Canc, Colon Canc Genet Grp, Edinburgh, Scotland..
    Farrington, Susan M.
    Univ Edinburgh, Med Res Council Inst Genet & Canc, Canc Res UK Edinburgh Ctr, Edinburgh, Scotland..
    Ding, Kefeng
    Zhejiang Univ, Affiliated Hosp 2, Colorectal Surg & Oncol, Key Lab Canc Prevent & Intervent,Minist Educ,Sch M, Hangzhou, Peoples R China..
    Theodoratou, Evropi
    Univ Edinburgh, Usher Inst, Ctr Global Hlth, Edinburgh, Scotland.;Univ Edinburgh, Med Res Council Inst Genet & Canc, Canc Res UK Edinburgh Ctr, Edinburgh, Scotland..
    Li, Xue
    Zhejiang Univ, Dept Big Data Hlth Sci, Sch Publ Hlth, Sch Med, Hangzhou, Zhejiang, Peoples R China.;Zhejiang Univ, Analyt Affiliated Hosp 2, Ctr Clin Big Data, Sch Med, Hangzhou, Zhejiang, Peoples R China.;Univ Edinburgh, Usher Inst, Ctr Global Hlth, Edinburgh, Scotland..
    Identification of novel protein biomarkers and drug targets for colorectal cancer by integrating human plasma proteome with genome2023In: Genome Medicine, E-ISSN 1756-994X, Vol. 15, no 1, article id 75Article in journal (Refereed)
    Abstract [en]

    Background: The proteome is a major source of therapeutic targets. We conducted a proteome-wide Mendelian randomization (MR) study to identify candidate protein markers and therapeutic targets for colorectal cancer (CRC).

    Methods: Protein quantitative trait loci (pQTLs) were derived from seven published genome-wide association studies (GWASs) on plasma proteome, and summary-level data were extracted for 4853 circulating protein markers. Genetic associations with CRC were obtained from a large-scale GWAS meta-analysis (16,871 cases and 26,328 controls), the FinnGen cohort (4957 cases and 304,197 controls), and the UK Biobank (9276 cases and 477,069 controls). Colocalization and summary-data-based MR (SMR) analyses were performed sequentially to verify the causal role of candidate proteins. Single cell-type expression analysis, protein-protein interaction (PPI), and druggability evaluation were further conducted to detect the specific cell type with enrichment expression and prioritize potential therapeutic targets.

    Results: Collectively, genetically predicted levels of 13 proteins were associated with CRC risk. Elevated levels of two proteins (GREM1, CHRDL2) and decreased levels of 11 proteins were associated with an increased risk of CRC, among which four (GREM1, CLSTN3, CSF2RA, CD86) were prioritized with the most convincing evidence. These protein-coding genes are mainly expressed in tissue stem cells, epithelial cells, and monocytes in colon tumor tissue. Two interactive pairs of proteins (GREM1 and CHRDL2; MMP2 and TIMP2) were identified to be involved in osteoclast differentiation and tumorigenesis pathways; four proteins (POLR2F, CSF2RA, CD86, MMP2) have been targeted for drug development on autoimmune diseases and other cancers, with the potentials of being repurposed as therapeutic targets for CRC.

    Conclusions: This study identified several protein biomarkers to be associated with CRC risk and provided new insights into the etiology and promising targets for the development of screening biomarkers and therapeutic drugs for CRC.

    Download full text (pdf)
    FULLTEXT01
  • 39.
    Sun, Song
    et al.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Biochemistry and Microbiology. Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Comp Sci, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada;.
    Weile, Jochen
    Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Comp Sci, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada.
    Verby, Marta
    Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada.
    Wu, Yingzhou
    Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Comp Sci, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada.
    Wang, Yang
    Dana Farber Canc Inst, CCSB, Boston, MA 02215 USA;Harvard Med Sch, Blavatnik Inst, Dept Genet, Boston, MA 02115 USA.
    Cote, Atina G.
    Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada.
    Fotiadou, Iosifina
    Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada.
    Kitaygorodsky, Julia
    Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada.
    Vidal, Marc
    Dana Farber Canc Inst, CCSB, Boston, MA 02215 USA;Harvard Med Sch, Blavatnik Inst, Dept Genet, Boston, MA 02115 USA.
    Rine, Jasper
    Univ Calif Berkeley, Calif Inst Quantitat Biosci, Berkeley, CA 94720 USA;Univ Calif Berkeley, Dept Mol & Cell Biol, 229 Stanley Hall, Berkeley, CA 94720 USA.
    Jesina, Pavel
    Charles Univ Prague, Fac Med 1, Dept Pediat & Adolescent Med, Prague 12808 2, Czech Republic;Gen Univ Hosp Prague, Prague 12808 2, Czech Republic.
    Kozich, Viktor
    Charles Univ Prague, Fac Med 1, Dept Pediat & Adolescent Med, Prague 12808 2, Czech Republic;Gen Univ Hosp Prague, Prague 12808 2, Czech Republic.
    Roth, Frederick P.
    Univ Toronto, Donnelly Ctr, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Mol Genet, Toronto, ON M5S 3E1, Canada;Univ Toronto, Dept Comp Sci, Toronto, ON M5S 3E1, Canada;Mt Sinai Hosp, Lunenfeld Tanenbaum Res Inst, Toronto, ON M5G 1X5, Canada.
    A proactive genotype-to-patient-phenotype map for cystathionine beta-synthase2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 13Article in journal (Refereed)
    Abstract [en]

    Background For the majority of rare clinical missense variants, pathogenicity status cannot currently be classified. Classical homocystinuria, characterized by elevated homocysteine in plasma and urine, is caused by variants in the cystathionine beta-synthase (CBS) gene, most of which are rare. With early detection, existing therapies are highly effective. Methods Damaging CBS variants can be detected based on their failure to restore growth in yeast cells lacking the yeast ortholog CYS4. This assay has only been applied reactively, after first observing a variant in patients. Using saturation codon-mutagenesis, en masse growth selection, and sequencing, we generated a comprehensive, proactive map of CBS missense variant function. Results Our CBS variant effect map far exceeds the performance of computational predictors of disease variants. Map scores correlated strongly with both disease severity (Spearman's rho = 0.9) and human clinical response to vitamin B-6 (rho = 0.93). Conclusions We demonstrate that highly multiplexed cell-based assays can yield proactive maps of variant function and patient response to therapy, even for rare variants not previously seen in the clinic.

    Download full text (pdf)
    FULLTEXT01
  • 40.
    Sánchez-Busó, Leonor
    et al.
    Centre for Genomic Pathogen Surveillance, Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK; Genomics and Health Area, Foundation for the Promotion of Health and Biomedical Research in the Valencian Community (FISABIO-Public Health), Valencia, Spain.
    Yeats, Corin A.
    Centre for Genomic Pathogen Surveillance, Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK.
    Taylor, Benjamin
    Centre for Genomic Pathogen Surveillance, Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK; Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, Cambridgeshire, UK.
    Goater, Richard J.
    Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, Cambridgeshire, UK; European Molecular Biology Lab, Heidelberg, Baden-Wuerttemberg, Germany.
    Underwood, Anthony
    Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, Cambridgeshire, UK.
    Abudahab, Khalil
    Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, Cambridgeshire, UK.
    Argimón, Silvia
    Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, Cambridgeshire, UK.
    Ma, Kevin C.
    Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
    Mortimer, Tatum D.
    Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA.
    Golparian, Daniel
    Örebro University, School of Medical Sciences. World Health Organization Collaborating Centre for Gonorrhoea and Other STIs, Department of Laboratory Medicine.
    Cole, Michelle J.
    National Infection Service, Public Health England, London, UK.
    Grad, Yonatan H.
    Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, MA, USA; Division of Infectious Diseases, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
    Martin, Irene
    National Microbiology Laboratory, Public Health Agency of Canada, Winnipeg, Manitoba, Canada.
    Raphael, Brian H.
    Division of STD prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA.
    Shafer, William M.
    Department of Microbiology and Immunology and Emory Antibiotic Resistance Center, Emory University School of Medicine, Atlanta, GA, USA; Laboratories of Bacterial Pathogenesis, Veterans Affairs Medical Center, Decatur, GA, USA.
    Town, Katy
    Division of STD prevention, National Center for HIV/AIDS, Viral Hepatitis, STD and TB Prevention, Centers for Disease Control and Prevention, Atlanta, GA, USA.
    Wi, Teodora
    Department of the Global HIV, Hepatitis and STI Programmes, World Health Organization, Geneva, Switzerland.
    Harris, Simon R.
    Microbiotica, Biodata Innovation Centre, Cambridge, Cambridgeshire, UK.
    Unemo, Magnus
    Örebro University, School of Medical Sciences. Örebro University Hospital. World Health Organization Collaborating Centre for Gonorrhoea and Other STIs, Department of Laboratory Medicine.
    Aanensen, David M.
    Centre for Genomic Pathogen Surveillance, Big Data Institute, Nuffield Department of Medicine, University of Oxford, Oxford, Oxfordshire, UK; Centre for Genomic Pathogen Surveillance, Wellcome Sanger Institute, Wellcome Genome Campus, Cambridge, Cambridgeshire, UK.
    A community-driven resource for genomic epidemiology and antimicrobial resistance prediction of Neisseria gonorrhoeae at Pathogenwatch2021In: Genome Medicine, E-ISSN 1756-994X, Vol. 13, no 1, article id 61Article in journal (Refereed)
    Abstract [en]

    BACKGROUND: Antimicrobial-resistant (AMR) Neisseria gonorrhoeae is an urgent threat to public health, as strains resistant to at least one of the two last-line antibiotics used in empiric therapy of gonorrhoea, ceftriaxone and azithromycin, have spread internationally. Whole genome sequencing (WGS) data can be used to identify new AMR clones and transmission networks and inform the development of point-of-care tests for antimicrobial susceptibility, novel antimicrobials and vaccines. Community-driven tools that provide an easy access to and analysis of genomic and epidemiological data is the way forward for public health surveillance.

    METHODS: Here we present a public health-focussed scheme for genomic epidemiology of N. gonorrhoeae at Pathogenwatch ( https://pathogen.watch/ngonorrhoeae ). An international advisory group of experts in epidemiology, public health, genetics and genomics of N. gonorrhoeae was convened to inform on the utility of current and future analytics in the platform. We implement backwards compatibility with MLST, NG-MAST and NG-STAR typing schemes as well as an exhaustive library of genetic AMR determinants linked to a genotypic prediction of resistance to eight antibiotics. A collection of over 12,000 N. gonorrhoeae genome sequences from public archives has been quality-checked, assembled and made public together with available metadata for contextualization.

    RESULTS: AMR prediction from genome data revealed specificity values over 99% for azithromycin, ciprofloxacin and ceftriaxone and sensitivity values around 99% for benzylpenicillin and tetracycline. A case study using the Pathogenwatch collection of N. gonorrhoeae public genomes showed the global expansion of an azithromycin-resistant lineage carrying a mosaic mtr over at least the last 10 years, emphasising the power of Pathogenwatch to explore and evaluate genomic epidemiology questions of public health concern.

    CONCLUSIONS: The N. gonorrhoeae scheme in Pathogenwatch provides customised bioinformatic pipelines guided by expert opinion that can be adapted to public health agencies and departments with little expertise in bioinformatics and lower-resourced settings with internet connection but limited computational infrastructure. The advisory group will assess and identify ongoing public health needs in the field of gonorrhoea, particularly regarding gonococcal AMR, in order to further enhance utility with modified or new analytic methods.

  • 41.
    Tang, Jing
    et al.
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Tan, Chong Yew
    Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
    Oresic, Matej
    VTT Technical Research Centre of Finland, Espoo, Finland.
    Vidal-Puig, Antonio
    Metabolic Research Laboratories, Institute of Metabolic Science, Addenbrooke's Hospital, Cambridge, UK.
    Integrating post-genomic approaches as a strategy to advance our understanding of health and disease2009In: Genome Medicine, E-ISSN 1756-994X, Vol. 1, no 3, article id 35Article, review/survey (Refereed)
    Abstract [en]

    Following the publication of the complete human genomic sequence, the post-genomic era is driven by the need to extract useful information from genomic data. Genomics, transcriptomics, proteomics, metabolomics, epidemiological data and microbial data provide different angles to our understanding of gene-environment interactions and the determinants of disease and health. Our goal and our challenge are to integrate these very different types of data and perspectives of disease into a global model suitable for dissecting the mechanisms of disease and for predicting novel therapeutic strategies. This review aims to highlight the need for and problems with complex data integration, and proposes a framework for data integration. While there are many obstacles to overcome, biological models based upon multiple datasets will probably become the basis that drives future biomedical research.

  • 42. Varemo, Leif
    et al.
    Henriksen, Tora Ida
    Scheele, Camilla
    Broholm, Christa
    Pedersen, Maria
    Uhlen, Mathias
    Pedersen, Bente Klarlund
    Nielsen, Jens
    KTH, School of Biotechnology (BIO), Gene Technology. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Type 2 diabetes and obesity induce similar transcriptional reprogramming in human myocytes2017In: Genome Medicine, E-ISSN 1756-994X, Vol. 9, article id 47Article in journal (Refereed)
    Abstract [en]

    Background: Skeletal muscle is one of the primary tissues involved in the development of type 2 diabetes (T2D). The close association between obesity and T2D makes it difficult to isolate specific effects attributed to the disease alone. Therefore, here we set out to identify and characterize intrinsic properties of myocytes, associated independently with T2D or obesity. Methods: We generated and analyzed RNA-seq data from primary differentiated myotubes from 24 human subjects, using a factorial design (healthy/T2D and non-obese/obese), to determine the influence of each specific factor on genome-wide transcription. This setup enabled us to identify intrinsic properties, originating from muscle precursor cells and retained in the corresponding myocytes. Bioinformatic and statistical methods, including differential expression analysis, gene-set analysis, and metabolic network analysis, were used to characterize the different myocytes. Results: We found that the transcriptional program associated with obesity alone was strikingly similar to that induced specifically by T2D. We identified a candidate epigenetic mechanism, H3K27me3 histone methylation, mediating these transcriptional signatures. T2D and obesity were independently associated with dysregulated myogenesis, down-regulated muscle function, and up-regulation of inflammation and extracellular matrix components. Metabolic network analysis identified that in T2D but not obesity a specific metabolite subnetwork involved in sphingolipid metabolism was transcriptionally regulated. Conclusions: Our findings identify inherent characteristics in myocytes, as a memory of the in vivo phenotype, without the influence from a diabetic or obese extracellular environment, highlighting their importance in the development of T2D.

  • 43.
    Voisin, Sarah
    et al.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Almén, Markus Sällman
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Biochemistry and Microbiology.
    Zheleznyakova, Galina Y.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Lundberg, Lina
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Zarei, Sanaz
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Castillo, Sandra
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Eriksson, Fia Ence
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Nilsson, Emil K.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Blueher, Matthias
    Univ Leipzig, IFB Adipos Dis, Fac Med, D-04103 Leipzig, Germany..
    Boettcher, Yvonne
    Univ Leipzig, IFB Adipos Dis, Fac Med, D-04103 Leipzig, Germany..
    Kovacs, Peter
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology. Univ Leipzig, IFB Adipos Dis, Fac Med, D-04103 Leipzig, Germany..
    Klovins, Janis
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology. Latvian Biomed Res & Study Ctr, LV-1067 Riga, Latvia..
    Rask-Andersen, Mathias
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Schiöth, Helgi B.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Neuroscience, Functional Pharmacology.
    Many obesity-associated SNPs strongly associate with DNA methylation changes at proximal promoters and enhancers2015In: Genome Medicine, E-ISSN 1756-994X, Vol. 7, article id 103Article in journal (Refereed)
    Abstract [en]

    Background: The mechanisms by which genetic variants, such as single nucleotide polymorphisms (SNPs), identified in genome-wide association studies act to influence body mass remain unknown for most of these SNPs, which continue to puzzle the scientific community. Recent evidence points to the epigenetic and chromatin states of the genome as having important roles. Methods: We genotyped 355 healthy young individuals for 52 known obesity-associated SNPs and obtained DNA methylation levels in their blood using the Illumina 450 K BeadChip. Associations between alleles and methylation at proximal cytosine residues were tested using a linear model adjusted for age, sex, weight category, and a proxy for blood cell type counts. For replication in other tissues, we used two open-access datasets (skin fibroblasts, n = 62; four brain regions, n = 121-133) and an additional dataset in subcutaneous and visceral fat (n = 149). Results: We found that alleles at 28 of these obesity-associated SNPs associate with methylation levels at 107 proximal CpG sites. Out of 107 CpG sites, 38 are located in gene promoters, including genes strongly implicated in obesity (MIR148A, BDNF, PTPMT1, NR1H3, MGAT1, SCGB3A1, HOXC12, PMAIP1, PSIP1, RPS10-NUDT3, RPS10, SKOR1, MAP2K5, SIX5, AGRN, IMMP1L, ELP4, ITIH4, SEMA3G, POMC, ADCY3, SSPN, LGR4, TUFM, MIR4721, SULT1A1, SULT1A2, APOBR, CLN3, SPNS1, SH2B1, ATXN2L, and IL27). Interestingly, the associated SNPs are in known eQTLs for some of these genes. We also found that the 107 CpGs are enriched in enhancers in peripheral blood mononuclear cells. Finally, our results indicate that some of these associations are not blood-specific as we successfully replicated four associations in skin fibroblasts. Conclusions: Our results strongly suggest that many obesity-associated SNPs are associated with proximal gene regulation, which was reflected by association of obesity risk allele genotypes with differential DNA methylation. This study highlights the importance of DNA methylation and other chromatin marks as a way to understand the molecular basis of genetic variants associated with human diseases and traits.

    Download full text (pdf)
    fulltext
  • 44.
    Wadelius, Mia
    et al.
    Uppsala University, Disciplinary Domain of Medicine and Pharmacy, Faculty of Medicine, Department of Medical Sciences, Clinical pharmacogenomics and osteoporosis.
    Alfirevic, Ana
    University of Liverpool.
    Pharmacogenomics and personalized medicine: the plunge into next-generation sequencing2011In: Genome Medicine, E-ISSN 1756-994X, Vol. 3, no 12, p. 78-Article in journal (Refereed)
    Abstract [en]

    A report on the 9th Annual Cold Spring Harbor/Wellcome Trust meeting 'Pharmacogenomics and Personalized Medicine', Hinxton, Cambridge, UK, 29 September to 2 October 2011.

  • 45.
    Wang, Xinan
    et al.
    Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, 667 Huntington Ave, MA, Boston, United States.
    Zhang, Ziwei
    Department of Medical Oncology, Dana-Farber Cancer Institute, MA, Boston, United States.
    Ding, Yi
    Bioinformatics Interdepartmental Program, University of California, Los Angeles, United States.
    Chen, Tony
    Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, MA, Boston, United States.
    Mucci, Lorelei
    Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, MA, Boston, United States.
    Albanes, Demetrios
    Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, MD, Bethesda, United States.
    Landi, Maria Teresa
    Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, MD, Bethesda, United States.
    Caporaso, Neil E.
    Division of Cancer Epidemiology and Genetics, National Cancer Institute, National Institutes of Health, MD, Bethesda, United States.
    Lam, Stephen
    Department of Medicine, British Columbia Cancer Agency, University of British Columbia, Vancouver, Canada.
    Tardon, Adonina
    Faculty of Medicine, University of Oviedo and CIBERESP, Oviedo, Spain.
    Chen, Chu
    Department of Epidemiology, University of Washington School of Public Health, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, WA, Seattle, United States.
    Bojesen, Stig E.
    Department of Clinical Biochemistry, Herlev and Gentofte Hospital, Copenhagen University Hospital, Copenhagen, Denmark.
    Johansson, Mattias
    Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France.
    Risch, Angela
    Department of Biosciences and Medical Biology, Allergy-Cancer-BioNano Research Centre, University of Salzburg, and Cancer Cluster Salzburg, Salzburg, Austria.
    Bickeböller, Heike
    Department of Genetic Epidemiology, University Medical Center, Georg August University Göttingen, Göttingen, Germany.
    Wichmann, H-Erich
    Institute of Medical Informatics, Biometry and Epidemiology, Ludwig Maximilians University, Munich, Germany.
    Rennert, Gadi
    Clalit National Cancer Control Center, Carmel Medical Center and Technion Faculty of Medicine, Carmel, Haifa, Israel.
    Arnold, Susanne
    Markey Cancer Center, University of Kentucky, KY, Lexington, United States.
    Brennan, Paul
    Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France.
    McKay, James D.
    Genomic Epidemiology Branch, International Agency for Research on Cancer (IARC/WHO), Lyon, France.
    Field, John K.
    Department of Molecular and Clinical Cancer Medicine, Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom.
    Shete, Sanjay S.
    Department of Biostatistics, The University of Texas MD Anderson Cancer Center, TX, Houston, United States.
    Le Marchand, Loic
    Epidemiology Program, University of Hawaii Cancer Center, HI, Honolulu, United States.
    Liu, Geoffrey
    Princess Margaret Cancer Centre, Dalla Lana School of Public Health, University of Toronto, Toronto, Canada.
    Andrew, Angeline S.
    Department of Epidemiology, Department of Community and Family Medicine, Dartmouth Geisel School of Medicine, NH, Hanover, United States.
    Kiemeney, Lambertus A.
    Department for Health Evidence, Department of Urology, Radboud University Medical Center, Nijmegen, Netherlands.
    Zienolddiny-Narui, Shan
    National Institute of Occupational Health, Oslo, Norway.
    Behndig, Annelie F.
    Umeå University, Faculty of Medicine, Department of Public Health and Clinical Medicine.
    Johansson, Mikael
    Umeå University, Faculty of Medicine, Department of Radiation Sciences.
    Cox, Angie
    Department of Oncology and Metabolism, The Medical School, University of Sheffield, Sheffield, United Kingdom.
    Lazarus, Philip
    Department of Pharmaceutical Sciences, College of Pharmacy, Washington State University, WA, Spokane, United States.
    Schabath, Matthew B.
    Department of Cancer Epidemiology, H. Lee Moffitt Cancer Center and Research Institute, FL, Tampa, United States.
    Aldrich, Melinda C.
    Department of Medicine, Department of Biomedical Informatics and Department of Thoracic Surgery, Vanderbilt University Medical Center, TN, Nashville, United States.
    Hung, Rayjean J.
    Lunenfeld-Tanenbaum Research Institute, Sinai Health System, Dalla Lana School of Public Health, University of Toronto, ON, Toronto, Canada.
    Amos, Christopher I.
    Institute for Clinical and Translational Research, Department of Medicine, Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, TX, Houston, United States.
    Lin, Xihong
    Department of Biostatistics, Harvard T.H. Chan School of Public Health, Harvard University, MA, Boston, United States.
    Christiani, David C.
    Department of Environmental Health, Harvard T.H. Chan School of Public Health, Harvard University, 667 Huntington Ave, MA, Boston, United States; Department of Epidemiology, Harvard T.H. Chan School of Public Health, Harvard University, MA, Boston, United States.
    Impact of individual level uncertainty of lung cancer polygenic risk score (PRS) on risk stratification2024In: Genome Medicine, E-ISSN 1756-994X, Vol. 16, no 1, article id 22Article in journal (Refereed)
    Abstract [en]

    Background: Although polygenic risk score (PRS) has emerged as a promising tool for predicting cancer risk from genome-wide association studies (GWAS), the individual-level accuracy of lung cancer PRS and the extent to which its impact on subsequent clinical applications remains largely unexplored.

    Methods: Lung cancer PRSs and confidence/credible interval (CI) were constructed using two statistical approaches for each individual: (1) the weighted sum of 16 GWAS-derived significant SNP loci and the CI through the bootstrapping method (PRS-16-CV) and (2) LDpred2 and the CI through posteriors sampling (PRS-Bayes), among 17,166 lung cancer cases and 12,894 controls with European ancestry from the International Lung Cancer Consortium. Individuals were classified into different genetic risk subgroups based on the relationship between their own PRS mean/PRS CI and the population level threshold.

    Results: Considerable variances in PRS point estimates at the individual level were observed for both methods, with an average standard deviation (s.d.) of 0.12 for PRS-16-CV and a much larger s.d. of 0.88 for PRS-Bayes. Using PRS-16-CV, only 25.0% of individuals with PRS point estimates in the lowest decile of PRS and 16.8% in the highest decile have their entire 95% CI fully contained in the lowest and highest decile, respectively, while PRS-Bayes was unable to find any eligible individuals. Only 19% of the individuals were concordantly identified as having high genetic risk (> 90th percentile) using the two PRS estimators. An increased relative risk of lung cancer comparing the highest PRS percentile to the lowest was observed when taking the CI into account (OR = 2.73, 95% CI: 2.12–3.50, P-value = 4.13 × 10−15) compared to using PRS-16-CV mean (OR = 2.23, 95% CI: 1.99–2.49, P-value = 5.70 × 10−46). Improved risk prediction performance with higher AUC was consistently observed in individuals identified by PRS-16-CV CI, and the best performance was achieved by incorporating age, gender, and detailed smoking pack-years (AUC: 0.73, 95% CI = 0.72–0.74). Conclusions: Lung cancer PRS estimates using different methods have modest correlations at the individual level, highlighting the importance of considering individual-level uncertainty when evaluating the practical utility of PRS.

    Download full text (pdf)
    fulltext
  • 46.
    Zhang, Huan
    et al.
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center of Paediatrics and Gynaecology and Obstetrics, H.K.H. Kronprinsessan Victorias barn- och ungdomssjukhus.
    Klareskog, Lars
    Karolinska Inst, Sweden.
    Matussek, Andreas
    Karolinska Univ Hosp Lab, Sweden.
    Pfister, Stefan M.
    Hopp Childrens Canc Ctr Heidelberg KiTZ, Germany; German Canc Res Ctr, Germany; German Canc Consortium DKTK, Germany; Heidelberg Univ Hosp, Germany.
    Benson, Mikael
    Linköping University, Department of Clinical and Experimental Medicine, Division of Children's and Women's health. Linköping University, Faculty of Medicine and Health Sciences. Region Östergötland, Center of Paediatrics and Gynaecology and Obstetrics, H.K.H. Kronprinsessan Victorias barn- och ungdomssjukhus.
    Editorial Material: Translating genomic medicine to the clinic: challenges and opportunities in GENOME MEDICINE, vol 11, issue , pp2019In: Genome Medicine, E-ISSN 1756-994X, Vol. 11, article id 9Article in journal (Other academic)
    Abstract [en]

    Editorial summaryGenomic medicine has considerable potential to provide novel diagnostic and therapeutic solutions for patients who have molecularly complex diseases and who are not responding to existing therapies. To bridge the gap between genomic medicine and clinical practice, integration of various data types, resources, and joint international initiatives will be required.

    Download full text (pdf)
    fulltext
  • 47.
    Zhong, Wen
    et al.
    KTH, Proteinvetenskap, Sweden.
    Gummesson, Anders
    Gothenburg Univ, Sahlgrenska Acad, Inst Med, Dept Mol & Clin Med, Gothenburg, Sweden.;Sahlgrens Univ Hosp, Dept Clin Genet & Genom, Gothenburg, Sweden.
    Abdellah, Tebani
    KTH, Proteinvetenskap, Sweden.
    Karlsson, Max
    KTH, Science for Life Laboratory, SciLifeLab, Sweden.
    Hong, Mun-Gwan
    KTH, Science for Life Laboratory, SciLifeLab, Sweden.
    Schwenk, Jochen M.
    KTH, Science for Life Laboratory, SciLifeLab, Sweden.
    Edfors, Fredrik
    KTH, Proteinvetenskap, Sweden.
    Bergström, Goran
    Gothenburg Univ, Sahlgrenska Acad, Inst Med, Dept Mol & Clin Med, Gothenburg, Sweden.;Sahlgrens Univ Hosp, Dept Clin Physiol, Gothenburg, Sweden.
    Fagerberg, Linn
    KTH, Science for Life Laboratory, SciLifeLab, Sweden.
    Uhlén, Mathias
    KTH, Science for Life Laboratory, SciLifeLab, Sweden.
    Whole-genome sequence association analysis of blood proteins in a longitudinal wellness cohort2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 53Article in journal (Refereed)
    Abstract [en]

    Background The human plasma proteome is important for many biological processes and targets for diagnostics and therapy. It is therefore of great interest to understand the interplay of genetic and environmental factors to determine the specific protein levels in individuals and to gain a deeper insight of the importance of genetic architecture related to the individual variability of plasma levels of proteins during adult life. Methods We have combined whole-genome sequencing, multiplex plasma protein profiling, and extensive clinical phenotyping in a longitudinal 2-year wellness study of 101 healthy individuals with repeated sampling. Analyses of genetic and non-genetic associations related to the variability of blood levels of proteins in these individuals were performed. Results The analyses showed that each individual has a unique protein profile, and we report on the intra-individual as well as inter-individual variation for 794 plasma proteins. A genome-wide association study (GWAS) using 7.3 million genetic variants identified by whole-genome sequencing revealed 144 independent variants across 107 proteins that showed strong association (P < 6 x 10(-11)) between genetics and the inter-individual variability on protein levels. Many proteins not reported before were identified (67 out of 107) with individual plasma level affected by genetics. Our longitudinal analysis further demonstrates that these levels are stable during the 2-year study period. The variability of protein profiles as a consequence of environmental factors was also analyzed with focus on the effects of weight loss and infections. Conclusions We show that the adult blood levels of many proteins are determined at birth by genetics, which is important for efforts aimed to understand the relationship between plasma proteome profiles and human biology and disease.

  • 48.
    Zhong, Wen
    et al.
    KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Gummesson, Anders
    Gothenburg Univ, Sahlgrenska Acad, Inst Med, Dept Mol & Clin Med, Gothenburg, Sweden.;Sahlgrens Univ Hosp, Dept Clin Genet & Genom, Gothenburg, Sweden..
    Abdellah, Tebani
    KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Karlsson, Max
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science.
    Hong, Mun-Gwan
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science.
    Schwenk, Jochen M.
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science.
    Edfors, Fredrik
    KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science. KTH, Centres, Science for Life Laboratory, SciLifeLab.
    Bergström, Goran
    Gothenburg Univ, Sahlgrenska Acad, Inst Med, Dept Mol & Clin Med, Gothenburg, Sweden.;Sahlgrens Univ Hosp, Dept Clin Physiol, Gothenburg, Sweden..
    Fagerberg, Linn
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science.
    Uhlén, Mathias
    KTH, Centres, Science for Life Laboratory, SciLifeLab. KTH, School of Engineering Sciences in Chemistry, Biotechnology and Health (CBH), Protein Science. Karolinska Inst, Dept Neurosci, Stockholm, Sweden..
    Whole-genome sequence association analysis of blood proteins in a longitudinal wellness cohort2020In: Genome Medicine, E-ISSN 1756-994X, Vol. 12, no 1, article id 53Article in journal (Refereed)
    Abstract [en]

    Background The human plasma proteome is important for many biological processes and targets for diagnostics and therapy. It is therefore of great interest to understand the interplay of genetic and environmental factors to determine the specific protein levels in individuals and to gain a deeper insight of the importance of genetic architecture related to the individual variability of plasma levels of proteins during adult life. Methods We have combined whole-genome sequencing, multiplex plasma protein profiling, and extensive clinical phenotyping in a longitudinal 2-year wellness study of 101 healthy individuals with repeated sampling. Analyses of genetic and non-genetic associations related to the variability of blood levels of proteins in these individuals were performed. Results The analyses showed that each individual has a unique protein profile, and we report on the intra-individual as well as inter-individual variation for 794 plasma proteins. A genome-wide association study (GWAS) using 7.3 million genetic variants identified by whole-genome sequencing revealed 144 independent variants across 107 proteins that showed strong association (P < 6 x 10(-11)) between genetics and the inter-individual variability on protein levels. Many proteins not reported before were identified (67 out of 107) with individual plasma level affected by genetics. Our longitudinal analysis further demonstrates that these levels are stable during the 2-year study period. The variability of protein profiles as a consequence of environmental factors was also analyzed with focus on the effects of weight loss and infections. Conclusions We show that the adult blood levels of many proteins are determined at birth by genetics, which is important for efforts aimed to understand the relationship between plasma proteome profiles and human biology and disease.

1 - 48 of 48
CiteExportLink to result list
Permanent link
Cite
Citation style
  • apa
  • ieee
  • modern-language-association-8th-edition
  • vancouver
  • Other style
More styles
Language
  • de-DE
  • en-GB
  • en-US
  • fi-FI
  • nn-NO
  • nn-NB
  • sv-SE
  • Other locale
More languages
Output format
  • html
  • text
  • asciidoc
  • rtf