Your browser doesn't support javascript.
loading
Clinical implementation of RNA sequencing for Mendelian disease diagnostics.
Yépez, Vicente A; Gusic, Mirjana; Kopajtich, Robert; Mertes, Christian; Smith, Nicholas H; Alston, Charlotte L; Ban, Rui; Beblo, Skadi; Berutti, Riccardo; Blessing, Holger; Ciara, Elzbieta; Distelmaier, Felix; Freisinger, Peter; Häberle, Johannes; Hayflick, Susan J; Hempel, Maja; Itkis, Yulia S; Kishita, Yoshihito; Klopstock, Thomas; Krylova, Tatiana D; Lamperti, Costanza; Lenz, Dominic; Makowski, Christine; Mosegaard, Signe; Müller, Michaela F; Muñoz-Pujol, Gerard; Nadel, Agnieszka; Ohtake, Akira; Okazaki, Yasushi; Procopio, Elena; Schwarzmayr, Thomas; Smet, Joél; Staufner, Christian; Stenton, Sarah L; Strom, Tim M; Terrile, Caterina; Tort, Frederic; Van Coster, Rudy; Vanlander, Arnaud; Wagner, Matias; Xu, Manting; Fang, Fang; Ghezzi, Daniele; Mayr, Johannes A; Piekutowska-Abramczuk, Dorota; Ribes, Antonia; Rötig, Agnès; Taylor, Robert W; Wortmann, Saskia B; Murayama, Kei.
Affiliation
  • Yépez VA; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
  • Gusic M; Department of Informatics, Technical University of Munich, Garching, Germany.
  • Kopajtich R; Quantitative Biosciences Munich, Department of Biochemistry, Ludwig-Maximilians-Universität, Munich, Germany.
  • Mertes C; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
  • Smith NH; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Alston CL; DZHK (German Centre for Cardiovascular Research), partner site Munich Heart Alliance, Munich, Germany.
  • Ban R; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
  • Beblo S; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Berutti R; Department of Informatics, Technical University of Munich, Garching, Germany.
  • Blessing H; Department of Informatics, Technical University of Munich, Garching, Germany.
  • Ciara E; Wellcome Centre for Mitochondrial Research, Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK.
  • Distelmaier F; NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK.
  • Freisinger P; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Häberle J; Department of Pediatric Neurology, Beijing Children's Hospital, Capital Medical University, National Center for Children's Health, Beijing, China.
  • Hayflick SJ; Department of Women and Child Health, Hospital for Children and Adolescents, Center for Pediatric Research Leipzig (CPL), Center for Rare Diseases, University Hospitals, University of Leipzig, Leipzig, Germany.
  • Hempel M; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
  • Itkis YS; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Kishita Y; Department for Inborn Metabolic Diseases, Children's and Adolescents' Hospital, University of Erlangen-Nürnberg, Erlangen, Germany.
  • Klopstock T; Department of Medical Genetics, Children's Memorial Health Institute, Warsaw, Poland.
  • Krylova TD; Department of General Pediatrics, Neonatology and Pediatric Cardiology, Heinrich-Heine-University, Düsseldorf, Germany.
  • Lamperti C; Department of Pediatrics, Klinikum Reutlingen, Reutlingen, Germany.
  • Lenz D; University Children's Hospital Zurich and Children's Research Centre, Zürich, Switzerland.
  • Makowski C; Department of Molecular and Medical Genetics, Oregon Health & Science University, Portland, USA.
  • Mosegaard S; Institute of Human Genetics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany.
  • Müller MF; Research Centre for Medical Genetics, Moscow, Russia.
  • Muñoz-Pujol G; Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Tokyo, Japan.
  • Nadel A; Department of Life Science, Faculty of Science and Engineering, Kindai University, Osaka, Japan.
  • Ohtake A; Department of Neurology, Friedrich-Baur-Institute, University Hospital, Ludwig-Maximilians-Universität, Munich, Germany.
  • Okazaki Y; German Center for Neurodegenerative Diseases (DZNE), Munich, Germany.
  • Procopio E; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany.
  • Schwarzmayr T; Research Centre for Medical Genetics, Moscow, Russia.
  • Smet J; Unit of Medical Genetics and Neurogenetics, Fondazione IRCCS (Istituto di Ricovero e Cura a Carattere Scientifico) Istituto Neurologico Carlo Besta, Milan, Italy.
  • Staufner C; Division of Neuropediatrics and Pediatric Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany.
  • Stenton SL; Department of Pediatrics, Technical University of Munich, Munich, Germany.
  • Strom TM; Research Unit for Molecular Medicine, Department of Clinical Medicine, Aarhus University, Aarhus, Denmark.
  • Terrile C; Department of Informatics, Technical University of Munich, Garching, Germany.
  • Tort F; Section of Inborn Errors of Metabolism-IBC, Department of Biochemistry and Molecular Genetics, Hospital Clínic, IDIBAPS, CIBERER, Barcelona, Spain.
  • Van Coster R; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
  • Vanlander A; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Wagner M; Department of Pediatrics & Clinical Genomics, Faculty of Medicine, Saitama Medical University, Saitama, Japan.
  • Xu M; Center for Intractable Diseases, Saitama Medical University Hospital, Saitama, Japan.
  • Fang F; Diagnostics and Therapeutics of Intractable Diseases, Intractable Disease Research Center, Juntendo University, Graduate School of Medicine, Tokyo, Japan.
  • Ghezzi D; Inborn Metabolic and Muscular Disorders Unit, Anna Meyer Children Hospital, Florence, Italy.
  • Mayr JA; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
  • Piekutowska-Abramczuk D; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Ribes A; Department of Pediatric Neurology and Metabolism, Ghent University Hospital, Ghent, Belgium.
  • Rötig A; Division of Neuropediatrics and Pediatric Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany.
  • Taylor RW; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
  • Wortmann SB; Institute of Neurogenomics, Helmholtz Zentrum München, Neuherberg, Germany.
  • Murayama K; Institute of Human Genetics, School of Medicine, Technical University of Munich, Munich, Germany.
Genome Med ; 14(1): 38, 2022 04 05.
Article in En | MEDLINE | ID: mdl-35379322
ABSTRACT

BACKGROUND:

Lack of functional evidence hampers variant interpretation, leaving a large proportion of individuals with a suspected Mendelian disorder without genetic diagnosis after whole genome or whole exome sequencing (WES). Research studies advocate to further sequence transcriptomes to directly and systematically probe gene expression defects. However, collection of additional biopsies and establishment of lab workflows, analytical pipelines, and defined concepts in clinical interpretation of aberrant gene expression are still needed for adopting RNA sequencing (RNA-seq) in routine diagnostics.

METHODS:

We implemented an automated RNA-seq protocol and a computational workflow with which we analyzed skin fibroblasts of 303 individuals with a suspected mitochondrial disease that previously underwent WES. We also assessed through simulations how aberrant expression and mono-allelic expression tests depend on RNA-seq coverage.

RESULTS:

We detected on average 12,500 genes per sample including around 60% of all disease genes-a coverage substantially higher than with whole blood, supporting the use of skin biopsies. We prioritized genes demonstrating aberrant expression, aberrant splicing, or mono-allelic expression. The pipeline required less than 1 week from sample preparation to result reporting and provided a median of eight disease-associated genes per patient for inspection. A genetic diagnosis was established for 16% of the 205 WES-inconclusive cases. Detection of aberrant expression was a major contributor to diagnosis including instances of 50% reduction, which, together with mono-allelic expression, allowed for the diagnosis of dominant disorders caused by haploinsufficiency. Moreover, calling aberrant splicing and variants from RNA-seq data enabled detecting and validating splice-disrupting variants, of which the majority fell outside WES-covered regions.

CONCLUSION:

Together, these results show that streamlined experimental and computational processes can accelerate the implementation of RNA-seq in routine diagnostics.
Subject(s)
Key words

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: RNA / Transcriptome Type of study: Diagnostic_studies / Guideline Limits: Humans Language: En Journal: Genome Med Year: 2022 Document type: Article Affiliation country:

Full text: 1 Collection: 01-internacional Database: MEDLINE Main subject: RNA / Transcriptome Type of study: Diagnostic_studies / Guideline Limits: Humans Language: En Journal: Genome Med Year: 2022 Document type: Article Affiliation country:
...