251
|
Videira G, Malaquias MJ, Laranjinha I, Martins R, Taipa R, Magalhães M. Diagnosis of Aicardi-Goutières Syndrome in Adults: A Case Series. Mov Disord Clin Pract 2020; 7:303-307. [PMID: 32258229 DOI: 10.1002/mdc3.12903] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Revised: 01/11/2020] [Accepted: 01/19/2020] [Indexed: 02/02/2023] Open
Abstract
Introduction Aicardi-Goutières syndrome (AGS) is a genetic disease presenting with early-onset encephalopathy, generalized dystonia, spasticity, and cognitive disability. Diagnosis may be difficult in adults, as the clinical course seems static from infancy. Methods AGS patients from an adult movement disorders outpatient clinic were retrospectively analyzed. Results A total of 5 patients and 1 asymptomatic carrier from 3 different families were identified. All had a homozygous c.529G>A,p.A177T mutation in exon 7 of the RNASEH2B gene. Two patients had neonatal-onset AGS, 2 had later onset forms, and 1 was slightly symptomatic. All were diagnosed in adulthood after chilblains, and basal ganglia calcifications were identified on computed tomography scans. Discussion AGS patients have marked phenotypic variability regarding psychomotor development and morbidity. The present series included 1 asymptomatic carrier and 1 slightly symptomatic patient, both with homozygous RNASEH2B mutations. Chilblains and basal ganglia calcifications identified on computed tomography scan (but not on magnetic resonance imaging) are important clues for late diagnosis.
Collapse
Affiliation(s)
- Gonçalo Videira
- Neurology Department Centro Hospitalar Universitário do Porto Porto Portugal
| | | | - Inês Laranjinha
- Neurology Department Centro Hospitalar Universitário do Porto Porto Portugal
| | - Ricardo Martins
- Neuroradiology Department Centro Hospitalar Universitário do Porto Porto Portugal
| | - Ricardo Taipa
- Neuropathology Department Centro Hospitalar Universitário do Porto Porto Portugal
| | - Marina Magalhães
- Neurology Department Centro Hospitalar Universitário do Porto Porto Portugal
| |
Collapse
|
252
|
Kellner V, Luke B. Molecular and physiological consequences of faulty eukaryotic ribonucleotide excision repair. EMBO J 2020; 39:e102309. [PMID: 31833079 PMCID: PMC6996501 DOI: 10.15252/embj.2019102309] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2019] [Revised: 10/22/2019] [Accepted: 11/26/2019] [Indexed: 01/11/2023] Open
Abstract
The duplication of the eukaryotic genome is an intricate process that has to be tightly safe-guarded. One of the most frequently occurring errors during DNA synthesis is the mis-insertion of a ribonucleotide instead of a deoxyribonucleotide. Ribonucleotide excision repair (RER) is initiated by RNase H2 and results in error-free removal of such mis-incorporated ribonucleotides. If left unrepaired, DNA-embedded ribonucleotides result in a variety of alterations within chromosomal DNA, which ultimately lead to genome instability. Here, we review how genomic ribonucleotides lead to chromosomal aberrations and discuss how the tight regulation of RER timing may be important for preventing unwanted DNA damage. We describe the structural impact of unrepaired ribonucleotides on DNA and chromatin and comment on the potential consequences for cellular fitness. In the context of the molecular mechanisms associated with faulty RER, we have placed an emphasis on how and why increased levels of genomic ribonucleotides are associated with severe autoimmune syndromes, neuropathology, and cancer. In addition, we discuss therapeutic directions that could be followed for pathologies associated with defective removal of ribonucleotides from double-stranded DNA.
Collapse
Affiliation(s)
- Vanessa Kellner
- Institute of Molecular Biology (IMB)MainzGermany
- Present address:
Department of BiologyNew York UniversityNew YorkNYUSA
| | - Brian Luke
- Institute of Molecular Biology (IMB)MainzGermany
- Institute of Developmental Biology and Neurobiology (IDN)Johannes Gutenberg UniversitätMainzGermany
| |
Collapse
|
253
|
Yu ZX, Song HM. Toward a better understanding of type I interferonopathies: a brief summary, update and beyond. World J Pediatr 2020; 16:44-51. [PMID: 31377974 DOI: 10.1007/s12519-019-00273-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2019] [Accepted: 05/24/2019] [Indexed: 12/17/2022]
Abstract
BACKGROUNDS Type I interferonopathy is a group of autoinflammatory disorders associated with prominent enhanced type I interferon signaling. The mechanisms are complex, and the clinical phenotypes are diverse. This review briefly summarized the recent progresses of type I interferonopathy focusing on the clinical and molecular features, pathogeneses, diagnoses and potential therapies. DATA SOURCES Original research articles and literature reviews published in PubMed-indexed journals. RESULTS Type I interferonopathies include Aicardi-Goutières syndrome, spondyloenchondro-dysplasia with immune dysregulation, stimulator of interferon genes-associated vasculopathy with onset in infancy, X-linked reticulate pigmentary disorder, ubiquitin-specific peptidase 18 deficiency, chronic atypical neutrophilic dermatitis with lipodystrophy, and Singleton-Merten syndrome originally. Other disorders including interferon-stimulated gene 15 deficiency and DNAse II deficiency are believed to be interferonopathies as well. Intracranial calcification, skin vasculopathy, interstitial lung disease, failure to thrive, skeletal development problems and autoimmune features are common. Abnormal responses to nucleic acid stimuli and defective regulation of protein degradation are main mechanisms in disease pathogenesis. First generation Janus kinase inhibitors including baricitinib, tofacitinib and ruxolitinib are useful for disease control. Reverse transcriptase inhibitors seem to be another option for Aicardi-Goutières syndrome. CONCLUSIONS Tremendous progress has been made for the discovery of type I interferonopathies and responsible genes. Janus kinase inhibitors and other agents have potential therapeutic roles. Future basic, translational and clinical studies towards disease monitoring and powerful therapies are warranted.
Collapse
Affiliation(s)
- Zhong-Xun Yu
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Hong-Mei Song
- Department of Pediatrics, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China.
| |
Collapse
|
254
|
Toward a better understanding of type I interferonopathies: a brief summary, update and beyond. WORLD JOURNAL OF PEDIATRICS : WJP 2020. [PMID: 31377974 DOI: 10.1007/s12519-019-00273-z)] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/04/2022]
Abstract
BACKGROUNDS Type I interferonopathy is a group of autoinflammatory disorders associated with prominent enhanced type I interferon signaling. The mechanisms are complex, and the clinical phenotypes are diverse. This review briefly summarized the recent progresses of type I interferonopathy focusing on the clinical and molecular features, pathogeneses, diagnoses and potential therapies. DATA SOURCES Original research articles and literature reviews published in PubMed-indexed journals. RESULTS Type I interferonopathies include Aicardi-Goutières syndrome, spondyloenchondro-dysplasia with immune dysregulation, stimulator of interferon genes-associated vasculopathy with onset in infancy, X-linked reticulate pigmentary disorder, ubiquitin-specific peptidase 18 deficiency, chronic atypical neutrophilic dermatitis with lipodystrophy, and Singleton-Merten syndrome originally. Other disorders including interferon-stimulated gene 15 deficiency and DNAse II deficiency are believed to be interferonopathies as well. Intracranial calcification, skin vasculopathy, interstitial lung disease, failure to thrive, skeletal development problems and autoimmune features are common. Abnormal responses to nucleic acid stimuli and defective regulation of protein degradation are main mechanisms in disease pathogenesis. First generation Janus kinase inhibitors including baricitinib, tofacitinib and ruxolitinib are useful for disease control. Reverse transcriptase inhibitors seem to be another option for Aicardi-Goutières syndrome. CONCLUSIONS Tremendous progress has been made for the discovery of type I interferonopathies and responsible genes. Janus kinase inhibitors and other agents have potential therapeutic roles. Future basic, translational and clinical studies towards disease monitoring and powerful therapies are warranted.
Collapse
|
255
|
Belot A, Rice GI, Omarjee SO, Rouchon Q, Smith EMD, Moreews M, Tusseau M, Frachette C, Bournhonesque R, Thielens N, Gaboriaud C, Rouvet I, Chopin E, Hoshino A, Latour S, Ranchin B, Cimaz R, Romagnani P, Malcus C, Fabien N, Sarda MN, Kassai B, Lega JC, Decramer S, Abou-Jaoude P, Bruce IN, Simonet T, Bardel C, Rollat-Farnier PA, Viel S, Reumaux H, O'Sullivan J, Walzer T, Mathieu AL, Marenne G, Ludwig T, Genin E, Ellingford J, Bader-Meunier B, Briggs TA, Beresford MW, Crow YJ. Contribution of rare and predicted pathogenic gene variants to childhood-onset lupus: a large, genetic panel analysis of British and French cohorts. THE LANCET. RHEUMATOLOGY 2020; 2:e99-e109. [PMID: 38263665 DOI: 10.1016/s2665-9913(19)30142-0] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 12/09/2019] [Accepted: 12/10/2019] [Indexed: 11/30/2022]
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) is a rare immunological disorder and genetic factors are considered important in its causation. Monogenic lupus has been associated with around 30 genotypes in humans and 60 in mice, while genome-wide association studies have identified more than 90 risk loci. We aimed to analyse the contribution of rare and predicted pathogenic gene variants in a population of unselected cases of childhood-onset SLE. METHODS For this genetic panel analysis we designed a next-generation sequencing panel comprising 147 genes, including all known lupus-causing genes in humans, and potentially lupus-causing genes identified through GWAS and animal models. We screened 117 probands fulfilling American College of Rheumatology (ACR) criteria for SLE, ascertained through British and French cohorts of childhood-onset SLE, and compared these data with those of 791 ethnically matched controls from the 1000 Genomes Project and 574 controls from the FREX Consortium. FINDINGS After filtering, mendelian genotypes were confirmed in eight probands, involving variants in C1QA, C1QC, C2, DNASE1L3, and IKZF1. Seven additional patients carried heterozygous variants in complement or type I interferon-associated autosomal recessive genes, with decreased concentrations of the encoded proteins C3 and C9 recorded in two patients. Rare variants that were predicted to be damaging were significantly enriched in the childhood-onset SLE cohort compared with controls; 25% of SLE probands versus 5% of controls were identified to harbour at least one rare, predicted damaging variant (p=2·98 × 10-11). Inborn errors of immunity were estimated to account for 7% of cases of childhood-onset SLE, with defects in innate immunity representing the main monogenic contribution. INTERPRETATION An accumulation of rare variants that are predicted to be damaging in SLE-associated genes might contribute to disease expression and clinical heterogeneity. FUNDING European Research Council.
Collapse
Affiliation(s)
- Alexandre Belot
- Paediatric Nephrology, Rheumatology, Dermatology Unit, Femme Mere Enfant Hospital, Hospices Civils de Lyon, France; CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France.
| | - Gillian I Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK
| | - Sulliman Ommar Omarjee
- CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Quentin Rouchon
- Data Mining and Modelling for Biomedicine, VIB Center for Inflammation Research, Ghent, Belgium; Department of Applied Mathematics, Computer Science and Statistics, Ghent University, Ghent, Belgium
| | - Eve M D Smith
- Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK; Department of Women and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Marion Moreews
- CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Maud Tusseau
- CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Cécile Frachette
- Paediatric Nephrology, Rheumatology, Dermatology Unit, Femme Mere Enfant Hospital, Hospices Civils de Lyon, France; CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Raphael Bournhonesque
- CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Nicole Thielens
- University of Grenoble Alpes, CNRS, CEA, IBS, F-38000 Grenoble, France
| | | | - Isabelle Rouvet
- Centre de biotechnologie cellulaire et Biothèque, Groupe Hospitalier Est, Hospices Civils de Lyon, 69677 Bron, France
| | - Emilie Chopin
- Centre de biotechnologie cellulaire et Biothèque, Groupe Hospitalier Est, Hospices Civils de Lyon, 69677 Bron, France
| | - Akihiro Hoshino
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France
| | - Sylvain Latour
- Laboratory of Lymphocyte Activation and Susceptibility to EBV infection, INSERM UMR 1163, Paris, France; University Paris Descartes Sorbonne Paris Cité, Imagine Institute, Paris, France
| | - Bruno Ranchin
- Paediatric Nephrology, Rheumatology, Dermatology Unit, Femme Mere Enfant Hospital, Hospices Civils de Lyon, France
| | - Rolando Cimaz
- Rheumatology Unit, Anna Meyer Children Hospital and University of Florence, University of Florence, Florence, Italy
| | - Paula Romagnani
- Nephrology Unit, Anna Meyer Children Hospital and University of Florence, University of Florence, Florence, Italy
| | - Christophe Malcus
- Service d'Immunologie, Hôpital Edouard Herriot, Hospices Civils de Lyon, 69437 Lyon, France
| | - Nicole Fabien
- Service d'immunologie, Hospices Civils de Lyon, CHLS, 69495 Pierre-Bénite, France
| | - Marie-Nathalie Sarda
- Service d'immunologie, Hospices Civils de Lyon, CHLS, 69495 Pierre-Bénite, France
| | - Behrouz Kassai
- EPICIME-CIC 1407 de Lyon, Inserm, Service de Pharmacotoxicologie, Hospices Civils de Lyon & Université Lyon 1, 69677, Bron, France
| | - Jean-Christophe Lega
- Internal Medicine Unit, CHLS, Hospices Civils de Lyon, Pierre Benite, Université de Lyon 1, Lyon, France
| | - Stéphane Decramer
- Department of Pediatric Nephrology, Centre Hospitalier Universitaire de Toulouse, Toulouse, France; Centre De Référence des Maladies Rénales Rares du Sud Ouest & Inserm U1048, France
| | - Pauline Abou-Jaoude
- Department of Paediatric Nephrology, St George Hospital, University Medical Center, Beirut, Lebanon
| | - Ian N Bruce
- Centre for Musculoskeletal Research, Division of Musculoskeletal and Dermatological Sciences, School of Biological Sciences, University of Manchester, Manchester, UK; NIHR Manchester Biomedical Research Centre, Manchester University NHS Foundation Trust, Manchester Academic Health Science Centre, University of Manchester, Manchester, UK
| | - Thomas Simonet
- Department of Biostatistics-bioinformatics, Hospices Civils de Lyon, 69677, Bron, France
| | - Claire Bardel
- Department of Biostatistics-bioinformatics, Hospices Civils de Lyon, 69677, Bron, France; CNRS UMR5558, Biometry and evolutionary biology lab, Lyon University, Lyon 1 University, F-69622 Villeurbanne, France
| | - Pierre Antoine Rollat-Farnier
- CNRS UMR5558, Biometry and evolutionary biology lab, Lyon University, Lyon 1 University, F-69622 Villeurbanne, France
| | - Sebastien Viel
- Service d'immunologie, Hospices Civils de Lyon, CHLS, 69495 Pierre-Bénite, France
| | | | - James O'Sullivan
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Thierry Walzer
- CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Anne-Laure Mathieu
- CIRI, Centre International de Recherche en Infectiologie/ International Center for Infectiology Research, Inserm, U1111, Ecole Normale Supérieure de Lyon, Université Lyon 1, CNRS, UMR5308, Lyon, France
| | - Gaelle Marenne
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France
| | - Thomas Ludwig
- Inserm, Univ Brest, EFS, UMR 1078, GGB, F-29200 Brest, France; CHU Brest, Brest, France
| | | | - Jamie Ellingford
- Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Brigitte Bader-Meunier
- Paediatric Rheumatology and Immunology Unit, Necker Hospital, Imagine Institution, Paris, France
| | - Tracy A Briggs
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, UK; Manchester Centre for Genomic Medicine, Manchester Academic Health Sciences Centre, St Mary's Hospital, Manchester University NHS Foundation Trust, Manchester, UK
| | - Michael W Beresford
- Paediatric Rheumatology, Alder Hey Children's NHS Foundation Trust, Liverpool, UK; Department of Women and Children's Health, Institute of Translational Medicine, University of Liverpool, Liverpool, UK
| | - Yanick J Crow
- Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Paris, France; Paris Descartes University, Sorbonne-Paris-Cité, Institut Imagine, Paris, France; Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
256
|
Cellucci T, Van Mater H, Graus F, Muscal E, Gallentine W, Klein-Gitelman MS, Benseler SM, Frankovich J, Gorman MP, Van Haren K, Dalmau J, Dale RC. Clinical approach to the diagnosis of autoimmune encephalitis in the pediatric patient. NEUROLOGY-NEUROIMMUNOLOGY & NEUROINFLAMMATION 2020; 7:7/2/e663. [PMID: 31953309 PMCID: PMC7051207 DOI: 10.1212/nxi.0000000000000663] [Citation(s) in RCA: 202] [Impact Index Per Article: 40.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Accepted: 11/25/2019] [Indexed: 12/21/2022]
Abstract
Objective Autoimmune encephalitis (AE) is an important and treatable cause of acute encephalitis. Diagnosis of AE in a developing child is challenging because of overlap in clinical presentations with other diseases and complexity of normal behavior changes. Existing diagnostic criteria for adult AE require modification to be applied to children, who differ from adults in their clinical presentations, paraclinical findings, autoantibody profiles, treatment response, and long-term outcomes. Methods A subcommittee of the Autoimmune Encephalitis International Working Group collaborated through conference calls and email correspondence to consider the pediatric-specific approach to AE. The subcommittee reviewed the literature of relevant AE studies and sought additional input from other expert clinicians and researchers. Results Existing consensus criteria for adult AE were refined for use in children. Provisional pediatric AE classification criteria and an algorithm to facilitate early diagnosis are proposed. There is also discussion about how to distinguish pediatric AE from conditions within the differential diagnosis. Conclusions Diagnosing AE is based on the combination of a clinical history consistent with pediatric AE and supportive diagnostic testing, which includes but is not dependent on antibody testing. The proposed criteria and algorithm require validation in prospective pediatric cohorts.
Collapse
Affiliation(s)
- Tania Cellucci
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Heather Van Mater
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Francesc Graus
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Eyal Muscal
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - William Gallentine
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Marisa S Klein-Gitelman
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Susanne M Benseler
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Jennifer Frankovich
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Mark P Gorman
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Keith Van Haren
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Josep Dalmau
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia
| | - Russell C Dale
- From the Department of Pediatrics (T.C.), McMaster Children's Hospital, McMaster University, Hamilton, Ontario, Canada; Department of Pediatrics (H.V.M.), Duke University, Durham, NC; Neuroimmunology Program (F.G., J.D.), Institut d'Investigacions Biomèdiques August Pi I Sunyer, Barcelona, Spain; Department of Pediatrics (E.M.), Baylor College of Medicine, Houston, TX; Department of Neurology (W.G., K.V.H.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Division of Rheumatology (M.S.K.-G.), Ann & Robert H, Lurie Children's Hospital of Chicago, Northwestern University Feinberg School of Medicine, IL; Department of Pediatrics (S.M.B.), Alberta Children's Hospital, University of Calgary, Canada; Division of Pediatric Allergy, Immunology and Rheumatology (J.F.), Lucile Packard Children's Hospital, Stanford University, Palo Alto, CA; Department of Neurology (M.P.G.), Boston Children's Hospital, Harvard University, MA; Department of Neurology (J.D.), University of Pennsylvania, Philadelphia; Catalan Institution for Research and Advanced Studies (ICREA) (J.D.); Neuroimmunology Group (R.C.D.), Children's Hospital at Westmead, Faculty of Medicine and Health, University of Sydney, New South Wales, Australia.
| |
Collapse
|
257
|
Lambe J, Murphy OC, Mu W, Sondergaard Schatz K, Barañano KW, Venkatesan A. Relapsing-remitting clinical course expands the phenotype of Aicardi-Goutières syndrome. Ann Clin Transl Neurol 2020; 7:254-258. [PMID: 31920009 PMCID: PMC7034496 DOI: 10.1002/acn3.50979] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 12/20/2019] [Accepted: 12/21/2019] [Indexed: 12/11/2022] Open
Abstract
Aicardi-Goutières syndrome (AGS) is a rare and likely underdiagnosed genetic leukoencephalopathy, typically presenting in infancy with encephalopathy and characteristic neuroimaging features, with residual static neurological deficits. We describe a patient who, following an initial presentation at the age of 12 months in keeping with AGS, exhibited a highly atypical relapsing course of neurological symptoms in adulthood with essentially normal neuroimaging. Whole-exome sequencing confirmed a pathogenic RNASEH2B gene variant consistent with AGS. This case highlights the expanding phenotypes associated with AGS and the potential role of whole-exome sequencing in facilitating an increase in the rate of diagnosis.
Collapse
Affiliation(s)
- Jeffrey Lambe
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Olwen C Murphy
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Weiyi Mu
- Department of Genetic Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | | | - Kristin W Barañano
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Arun Venkatesan
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
258
|
Gacem N, Kavo A, Zerad L, Richard L, Mathis S, Kapur RP, Parisot M, Amiel J, Dufour S, de la Grange P, Pingault V, Vallat JM, Bondurand N. ADAR1 mediated regulation of neural crest derived melanocytes and Schwann cell development. Nat Commun 2020; 11:198. [PMID: 31924792 PMCID: PMC6954203 DOI: 10.1038/s41467-019-14090-5] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2019] [Accepted: 12/09/2019] [Indexed: 01/14/2023] Open
Abstract
The neural crest gives rise to numerous cell types, dysfunction of which contributes to many disorders. Here, we report that adenosine deaminase acting on RNA (ADAR1), responsible for adenosine-to-inosine editing of RNA, is required for regulating the development of two neural crest derivatives: melanocytes and Schwann cells. Neural crest specific conditional deletion of Adar1 in mice leads to global depigmentation and absence of myelin from peripheral nerves, resulting from alterations in melanocyte survival and differentiation of Schwann cells, respectively. Upregulation of interferon stimulated genes precedes these defects, which are associated with the triggering of a signature resembling response to injury in peripheral nerves. Simultaneous extinction of MDA5, a key sensor of unedited RNA, rescues both melanocytes and myelin defects in vitro, suggesting that ADAR1 safeguards neural crest derivatives from aberrant MDA5-mediated interferon production. We thus extend the landscape of ADAR1 function to the fields of neural crest development and disease.
Collapse
Affiliation(s)
- Nadjet Gacem
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Universite Paris Descartes-Universite de Paris, Paris, France.,INSERM, U955, Equipe 06, 8, rue du General Sarrail, 94010, Creteil Cedex, France
| | - Anthula Kavo
- INSERM, U955, Equipe 06, 8, rue du General Sarrail, 94010, Creteil Cedex, France.,Faculte de Medecine, Universite Paris Est, 94000, Creteil, France
| | - Lisa Zerad
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Universite Paris Descartes-Universite de Paris, Paris, France
| | - Laurence Richard
- Department of Neurology, Centre de Reference Neuropathies Peripheriques Rares, 2 avenue Martin-Luther-King, 87042, Limoges, France
| | - Stephane Mathis
- Department of Neurology (Nerve-Muscle Unit) and Grand Sud-Ouest National Reference Center for Neuromuscular Disorders, CHU Bordeaux, Pellegrin Hospital, 33076, Bordeaux, France
| | - Raj P Kapur
- Department of Pathology, Seattle Children's Hospital and University of Washington, 4800 Sand Point Way NE, Seattle, WA, 98105, USA
| | - Melanie Parisot
- Genomics Core Facility, Institut Imagine-Structure Federative de Recherche Necker, INSERM U1163 and INSERM US24/CNRS UMS3633, 24 bvd Montparnasse, 75015, Paris, France
| | - Jeanne Amiel
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Universite Paris Descartes-Universite de Paris, Paris, France
| | - Sylvie Dufour
- INSERM, U955, Equipe 06, 8, rue du General Sarrail, 94010, Creteil Cedex, France.,Faculte de Medecine, Universite Paris Est, 94000, Creteil, France
| | | | - Veronique Pingault
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Universite Paris Descartes-Universite de Paris, Paris, France.,Service de Genetique Moleculaire, Hopital Necker-Enfants-Malades, 149 rue de Sevres, 75015, Paris, France
| | - Jean Michel Vallat
- Department of Neurology, Centre de Reference Neuropathies Peripheriques Rares, 2 avenue Martin-Luther-King, 87042, Limoges, France
| | - Nadege Bondurand
- Laboratory of Embryology and Genetics of Human Malformation, Imagine Institute, INSERM UMR 1163, Universite Paris Descartes-Universite de Paris, Paris, France. .,INSERM, U955, Equipe 06, 8, rue du General Sarrail, 94010, Creteil Cedex, France.
| |
Collapse
|
259
|
Tonduti D, Fazzi E, Badolato R, Orcesi S. Novel and emerging treatments for Aicardi-Goutières syndrome. Expert Rev Clin Immunol 2020; 16:189-198. [PMID: 31855085 DOI: 10.1080/1744666x.2019.1707663] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Introduction: Aicardi-Goutières syndrome (AGS) is the prototype of the type I interferonopathies, a new heterogeneous group of autoinflammatory disorders in which type I interferon plays a pivotal role. The disease usually manifests itself during infancy, primarily affecting the brain and the skin, and is characterized by cerebrospinal fluid chronic lymphocytosis and raised levels of interferon-alpha and by cardinal neuroradiological features: cerebral calcification, leukoencephalopathy and cerebral atrophy. Recently many aspects of the pathogenesis of AGS have been clarified, making it possible to hypothesize new therapeutic strategies.Areas covered: We here review recent data concerning pathogenesis and novel therapeutic strategies in AGS, including the use of Janus kinase inhibitors, reverse transcriptase inhibitors, anti-IFN-α antibodies, anti-interleukin antibodies, antimalarial drugs and other cGAS inhibitors.Expert opinion: Thanks to the identification of the molecular basis of AGS, many aspects of its pathogenesis have been clarified, making it possible to propose new therapeutic strategies for AGS and type I interferonopathies. A number of therapeutic options are now becoming possible, even though their efficacy is still to be proven. However, in spite of research advances coming from clinical trials and case series, there are still a number of open questions, which urgently need to be addressed.
Collapse
Affiliation(s)
- Davide Tonduti
- Paediatric Neurology Unit, V. Buzzi Children's Hospital, Milan, Italy
| | - Elisa Fazzi
- Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy.,Unit of Child Neurology and Psychiatry, ASST Spedali Civili of Brescia, Brescia, Italy
| | - Raffaele Badolato
- Molecular Medicine Institute "Angelo Nocivelli" and Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Simona Orcesi
- Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy.,Unit of Child and Adolescent Neurology, IRCCS Mondino Foundation, Pavia, Italy
| |
Collapse
|
260
|
Arbustini E, Narula N, Giuliani L, Di Toro A. Genetic Basis of Myocarditis: Myth or Reality? MYOCARDITIS 2020. [PMCID: PMC7122345 DOI: 10.1007/978-3-030-35276-9_4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The genetic basis of myocarditis remains an intriguing concept, at least as long as the definition of myocarditis constitutes the definitive presence of myocardial inflammation sufficient to cause the observed ventricular dysfunction in the setting of cardiotropic infections. Autoimmune or immune-mediated myocardial inflammation constitutes a complex area of clinical interest, wherein numerous and not yet fully understood role of hereditary auto-inflammatory diseases can result in inflammation of the pericardium and myocardium. Finally, myocardial involvement in hereditary immunodeficiency diseases, cellular and humoral, is a possible trigger for infections which may complicate the diseases themselves. Whether the role of constitutional genetics can make the patient susceptible to myocardial inflammation remains yet to be explored.
Collapse
|
261
|
Adang L, Gavazzi F, De Simone M, Fazzi E, Galli J, Koh J, Kramer-Golinkoff J, De Giorgis V, Orcesi S, Peer K, Ulrick N, Woidill S, Shults J, Vanderver A. Developmental Outcomes of Aicardi Goutières Syndrome. J Child Neurol 2020; 35:7-16. [PMID: 31559893 PMCID: PMC7402202 DOI: 10.1177/0883073819870944] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Aicardi Goutières syndrome is a monogenic interferonopathy caused by abnormalities in the intracellular nucleic acid sensing machinery (TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR1, or IFIH1). Most individuals affected by Aicardi Goutières syndrome exhibit some degree of neurologic impairment, from spastic paraparesis with relatively preserved cognition to tetraparesis and severe intellectual disability. Because of this heterogeneity, it is important to fully characterize the developmental trajectory in Aicardi Goutières syndrome. To characterize the clinical presentation in Aicardi Goutières syndrome, early features were collected from an international cohort of children (n = 100) with genetically confirmed Aicardi Goutières syndrome. There was a heterogeneous age of onset, with overlapping clusters of presenting symptoms: altered mental status, systemic inflammatory symptoms, and acute neurologic disability. Next, we created genotype-specific developmental milestone acquisition curves. Individuals with microcephaly or TREX1-related Aicardi Goutières syndrome secondary were the most severely affected and less likely to reach milestones, including head control, sitting, and nonspecific mama/dada. Individuals affected by SAMHD1, IFIH1, and ADAR attained the most advanced milestones, with 44% achieving verbal communication and 31% independently ambulating. Retrospective function scales (Gross Motor Function Classification System, Manual Ability Classification System, and Communication Function Classification System) demonstrated that two-thirds of the Aicardi Goutières syndrome population are severely affected. Our results suggest multifactorial influences on developmental trajectory, including a strong contribution from genotype. Further studies are needed to identify the additional factors that influence overall outcomes to better counsel families and to design clinical trials with appropriate clinical endpoints.
Collapse
Affiliation(s)
- Laura Adang
- Division of Neurology, Children’s Hospital of Philadelphia
| | | | | | - Elisa Fazzi
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia,Department of Clinical and Experimental Sciences, University of Brescia
| | - Jessica Galli
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia,Department of Clinical and Experimental Sciences, University of Brescia
| | - Jamie Koh
- Division of Neurology, Children’s Hospital of Philadelphia
| | | | | | - Simona Orcesi
- Child Neurology and Psychiatry Unit, IRCCS Mondino Foundation, Pavia, Italy,Department of Brain and Behavioral Sciences, University of Pavia, Pavia, Italy
| | - Kyle Peer
- Division of Neurology, Children’s Hospital of Philadelphia
| | - Nicole Ulrick
- Division of Neurology, Children’s Hospital of Philadelphia
| | - Sarah Woidill
- Division of Neurology, Children’s Hospital of Philadelphia
| | - Justine Shults
- Department of Biostatistics. Perelman School of Medicine at University of Pennsylvania
| | | |
Collapse
|
262
|
Crow YJ, Shetty J, Livingston JH. Treatments in Aicardi-Goutières syndrome. Dev Med Child Neurol 2020; 62:42-47. [PMID: 31175662 DOI: 10.1111/dmcn.14268] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 04/11/2019] [Indexed: 12/25/2022]
Abstract
Comprehensive reviews of the clinical characteristics and pathogenesis of Aicardi-Goutières syndrome (AGS), particularly its contextualization within a putative type I interferonopathy framework, already exist. However, recent reports of attempts at treatment suggest that an assessment of the field from a therapeutic perspective is warranted at this time. Here, we briefly summarize the neurological phenotypes associated with mutations in the seven genes so far associated with AGS, rehearse current knowledge of the pathology as it relates to possible treatment approaches, critically appraise the potential utility of therapies, and discuss the challenges in assessing clinical efficacy. WHAT THIS PAPER ADDS: Progress in understanding AGS disease pathogenesis has led to the first attempts at targeted treatment. Further rational therapies are expected to become available in the short- to medium-term.
Collapse
Affiliation(s)
- Yanick J Crow
- Centre for Genomic and Experimental Medicine, MRC Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK.,Laboratory of Neurogenetics and Neuroinflammation, Institut Imagine, Paris Descartes University, Paris, France
| | - Jayakara Shetty
- Royal Hospital for Sick Children, NHS Lothian, Edinburgh, UK.,Department of Child Life and Health, MRC Centre for Reproductive Health, University of Edinburgh, Edinburgh, UK
| | - John H Livingston
- Department of Paediatric Neurology, Leeds General Infirmary, Leeds, UK
| |
Collapse
|
263
|
Nishimura T, Baba M, Ogawa S, Kojima K, Takita T, Crouch RJ, Yasukawa K. Characterization of six recombinant human RNase H2 bearing Aicardi-Goutiéres syndrome causing mutations. J Biochem 2019; 166:537-545. [PMID: 31529068 DOI: 10.1093/jb/mvz073] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 09/02/2019] [Indexed: 01/18/2023] Open
Abstract
Mammalian RNase H2 is a heterotrimeric enzyme consisting of one catalytic subunit (A) and two accessory subunits (B and C). RNase H2 is involved in the removal of a single ribonucleotide embedded in genomic DNA and removal of RNA of RNA/DNA hybrids. In humans, mutation of the RNase H2 gene causes a severe neuroinflammatory disorder Aicardi-Goutières syndrome (AGS). Here, we examined the activity and stability of six recombinant human RNase H2 variants bearing one AGS-causing mutation, A-G37S (Gly37 in the A subunit is replaced with Ser), A-N212I, A-R291H, B-A177T, B-V185G, or C-R69W. The activity of A-G37S was 0.3-1% of that of the wild-type RNase H2 (WT), while those of other five variants were 51-120%. In circular dichroism measurement, the melting temperatures of variants were 50-53°C, lower than that of WT (56°C). These results suggested that A-G37S had decreased activity and stability than WT, while other five variants had decreased stability but retained activity. In gel filtration chromatography of the purified enzyme preparation, WT migrated as a heterotrimer, while A-R291H eluted in two separate peaks containing either the heterotrimer or only the A subunit, suggesting that some AGS-causing mutations affect the heterotrimer-forming stability of RNase H2.
Collapse
Affiliation(s)
- Takuto Nishimura
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Oiwakecho, Kitashirakawa, Sakyoku, Kyoto 606-8502, Japan
| | - Misato Baba
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Oiwakecho, Kitashirakawa, Sakyoku, Kyoto 606-8502, Japan
| | - Saori Ogawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Oiwakecho, Kitashirakawa, Sakyoku, Kyoto 606-8502, Japan
| | - Kenji Kojima
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Oiwakecho, Kitashirakawa, Sakyoku, Kyoto 606-8502, Japan
| | - Teisuke Takita
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Oiwakecho, Kitashirakawa, Sakyoku, Kyoto 606-8502, Japan
| | - Robert J Crouch
- Section on Formation of RNA, Division of Developmental Biology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD 20892, USA
| | - Kiyoshi Yasukawa
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Oiwakecho, Kitashirakawa, Sakyoku, Kyoto 606-8502, Japan
| |
Collapse
|
264
|
Demirkaya E, Arici ZS, Romano M, Berard RA, Aksentijevich I. Current State of Precision Medicine in Primary Systemic Vasculitides. Front Immunol 2019; 10:2813. [PMID: 31921111 PMCID: PMC6927998 DOI: 10.3389/fimmu.2019.02813] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 11/15/2019] [Indexed: 12/12/2022] Open
Abstract
Precision medicine (PM) is an emerging data-driven health care approach that integrates phenotypic, genomic, epigenetic, and environmental factors unique to an individual. The goal of PM is to facilitate diagnosis, predict effective therapy, and avoid adverse reactions specific for each patient. The forefront of PM is in oncology; nonetheless, it is developing in other fields of medicine, including rheumatology. Recent studies on elucidating the genetic architecture of polygenic and monogenic rheumatological diseases have made PM possible by enabling physicians to customize medical treatment through the incorporation of clinical features and genetic data. For complex inflammatory disorders, the prevailing paradigm is that disease susceptibility is due to additive effects of common reduced-penetrance gene variants and environmental factors. Efforts have been made to calculate cumulative genetic risk score (GRS) and to relate specific susceptibility alleles for use of target therapies. The discovery of rare patients with single-gene high-penetrance mutations informed our understanding of pathways driving systemic inflammation. Here, we review the advances in practicing PM in patients with primary systemic vasculitides (PSVs). We summarize recent genetic studies and discuss current knowledge on the contribution of epigenetic factors and extracellular vesicles (EVs) in disease progression and treatment response. Implementation of PM in PSVs is a developing field that will require analysis of a large cohort of patients to validate data from genomics, transcriptomics, metabolomics, proteomics, and epigenomics studies for accurate disease profiling. This multi-omics approach to study disease pathogeneses should ultimately provide a powerful tool for stratification of patients to receive tailored optimal therapies and for monitoring their disease activity.
Collapse
Affiliation(s)
- Erkan Demirkaya
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Zehra Serap Arici
- Department of Paediatric Rheumatology, Sanliurfa Training and Research Hospital, Sanliurfa, Turkey
| | - Micol Romano
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada.,Department of Pediatric Rheumatology, Istituto Ortopedico Gaetano Pini, Milan, Italy
| | - Roberta Audrey Berard
- Division of Paediatric Rheumatology, Department of Paediatrics, Schulich School of Medicine & Dentistry, University of Western Ontario, London, ON, Canada
| | - Ivona Aksentijevich
- Inflammatory Disease Section, National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
265
|
Ashrafi MR, Amanat M, Garshasbi M, Kameli R, Nilipour Y, Heidari M, Rezaei Z, Tavasoli AR. An update on clinical, pathological, diagnostic, and therapeutic perspectives of childhood leukodystrophies. Expert Rev Neurother 2019; 20:65-84. [PMID: 31829048 DOI: 10.1080/14737175.2020.1699060] [Citation(s) in RCA: 44] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Introduction: Leukodystrophies constitute heterogenous group of rare heritable disorders primarily affecting the white matter of central nervous system. These conditions are often under-appreciated among physicians. The first clinical manifestations of leukodystrophies are often nonspecific and can occur in different ages from neonatal to late adulthood periods. The diagnosis is, therefore, challenging in most cases.Area covered: Herein, the authors discuss different aspects of leukodystrophies. The authors used MEDLINE, EMBASE, and GOOGLE SCHOLAR to provide an extensive update about epidemiology, classifications, pathology, clinical findings, diagnostic tools, and treatments of leukodystrophies. Comprehensive evaluation of clinical findings, brain magnetic resonance imaging, and genetic studies play the key roles in the early diagnosis of individuals with leukodystrophies. No cure is available for most heritable white matter disorders but symptomatic treatments can significantly decrease the burden of events. New genetic methods and stem cell transplantation are also under investigation to further increase the quality and duration of life in affected population.Expert opinion: The improvements in molecular diagnostic tools allow us to identify the meticulous underlying etiology of leukodystrophies and result in higher diagnostic rates, new classifications of leukodystrophies based on genetic information, and replacement of symptomatic managements with more specific targeted therapies.Abbreviations: 4H: Hypomyelination, hypogonadotropic hypogonadism and hypodontia; AAV: Adeno-associated virus; AD: autosomal dominant; AGS: Aicardi-Goutieres syndrome; ALSP: Axonal spheroids and pigmented glia; APGBD: Adult polyglucosan body disease; AR: autosomal recessive; ASO: Antisense oligonucleotide therapy; AxD: Alexander disease; BAEP: Brainstem auditory evoked potentials; CAA: Cerebral amyloid angiopathy; CADASIL: Cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy; CARASAL: Cathepsin A-related arteriopathy with strokes and leukoencephalopathy; CARASIL: Cerebral autosomal recessive arteriopathy with subcortical infarcts and leukoencephalopathy; CGH: Comparative genomic hybridization; ClC2: Chloride Ion Channel 2; CMTX: Charcot-Marie-Tooth disease, X-linked; CMV: Cytomegalovirus; CNS: central nervous system; CRISP/Cas9: Clustered regularly interspaced short palindromic repeat/CRISPR-associated 9; gRNA: Guide RNA; CTX: Cerebrotendinous xanthomatosis; DNA: Deoxyribonucleic acid; DSB: Double strand breaks; DTI: Diffusion tensor imaging; FLAIR: Fluid attenuated inversion recovery; GAN: Giant axonal neuropathy; H-ABC: Hypomyelination with atrophy of basal ganglia and cerebellum; HBSL: Hypomyelination with brainstem and spinal cord involvement and leg spasticity; HCC: Hypomyelination with congenital cataracts; HEMS: Hypomyelination of early myelinated structures; HMG CoA: Hydroxy methylglutaryl CoA; HSCT: Hematopoietic stem cell transplant; iPSC: Induced pluripotent stem cells; KSS: Kearns-Sayre syndrome; L-2-HGA: L-2-hydroxy glutaric aciduria; LBSL: Leukoencephalopathy with brainstem and spinal cord involvement and elevated lactate; LCC: Leukoencephalopathy with calcifications and cysts; LTBL: Leukoencephalopathy with thalamus and brainstem involvement and high lactate; MELAS: Mitochondrial myopathy, encephalopathy, lactic acidosis, and stroke; MERRF: Myoclonic epilepsy with ragged red fibers; MLC: Megalencephalic leukoencephalopathy with subcortical cysts; MLD: metachromatic leukodystrophy; MRI: magnetic resonance imaging; NCL: Neuronal ceroid lipofuscinosis; NGS: Next generation sequencing; ODDD: Oculodentodigital dysplasia; PCWH: Peripheral demyelinating neuropathy-central-dysmyelinating leukodystrophy-Waardenburg syndrome-Hirschprung disease; PMD: Pelizaeus-Merzbacher disease; PMDL: Pelizaeus-Merzbacher-like disease; RNA: Ribonucleic acid; TW: T-weighted; VWM: Vanishing white matter; WES: whole exome sequencing; WGS: whole genome sequencing; X-ALD: X-linked adrenoleukodystrophy; XLD: X-linked dominant; XLR: X-linked recessive.
Collapse
Affiliation(s)
- Mahmoud Reza Ashrafi
- Myelin Disorders Clinic, Department of Pediatric Neurology, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Man Amanat
- Faculty of Medicine, Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Masoud Garshasbi
- Department of Medical Genetics, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Reyhaneh Kameli
- Myelin Disorders Clinic, Department of Pediatric Neurology, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Yalda Nilipour
- Pediatric pathology research center, research institute for children's health, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Morteza Heidari
- Myelin Disorders Clinic, Department of Pediatric Neurology, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Rezaei
- Myelin Disorders Clinic, Department of Pediatric Neurology, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| | - Ali Reza Tavasoli
- Myelin Disorders Clinic, Department of Pediatric Neurology, Children's Medical Center, Pediatrics Center of Excellence, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
266
|
Uehara R, Cerritelli SM, Hasin N, Sakhuja K, London M, Iranzo J, Chon H, Grinberg A, Crouch RJ. Two RNase H2 Mutants with Differential rNMP Processing Activity Reveal a Threshold of Ribonucleotide Tolerance for Embryonic Development. Cell Rep 2019; 25:1135-1145.e5. [PMID: 30380406 PMCID: PMC6309994 DOI: 10.1016/j.celrep.2018.10.019] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 09/18/2018] [Accepted: 10/03/2018] [Indexed: 11/30/2022] Open
Abstract
RNase H2 has two distinct functions: initiation of the ribonucleotide excision repair (RER) pathway by cleaving ribonucleotides (rNMPs) incorporated during DNA replication and processing the RNA portion of an R-loop formed during transcription. An RNase H2 mutant lacking RER activity but supporting R-loop removal revealed that rNMPs in DNA initiate p53-dependent DNA damage response and early embryonic arrest in mouse. However, an RNase H2 AGS-related mutant with residual RER activity develops to birth. Estimations of the number of rNMPs in DNA in these two mutants define a ribonucleotide threshold above which p53 induces apoptosis. Below the threshold, rNMPs in DNA trigger an innate immune response. Compound heterozygous cells, containing both defective enzymes, retain rNMPs above the threshold, indicative of competition for RER substrates between active and inactive enzymes, suggesting that patients with compound heterozygous mutations in RNASEH2 genes may not reflect the properties of recombinantly expressed proteins. Uehara et al. use RNase H2 mice with differing activity levels for removal of rNMPs embedded in DNA. Moderate levels of rNMPs lead to perinatal lethality activating the cGAS-Sting DNA sensing innate immune response. Exceeding a threshold, high abundance of rNMPs activates p53-dependent DNA damage, causing early embryonic lethality.
Collapse
Affiliation(s)
- Ryo Uehara
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Susana M Cerritelli
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Naushaba Hasin
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Kiran Sakhuja
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Mariya London
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Jaime Iranzo
- NCBI, National Library of Medicine, Bethesda, MD, USA
| | - Hyongi Chon
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Alexander Grinberg
- Mouse Core, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA
| | - Robert J Crouch
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, MD, USA.
| |
Collapse
|
267
|
The pathogenesis of systemic lupus erythematosus: Harnessing big data to understand the molecular basis of lupus. J Autoimmun 2019; 110:102359. [PMID: 31806421 DOI: 10.1016/j.jaut.2019.102359] [Citation(s) in RCA: 80] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2019] [Accepted: 11/04/2019] [Indexed: 12/22/2022]
Abstract
Systemic lupus erythematosus (SLE) is a chronic, systemic autoimmune disease that causes damage to multiple organ systems. Despite decades of research and available murine models that capture some aspects of the human disease, new treatments for SLE lag behind other autoimmune diseases such as Rheumatoid Arthritis and Crohn's disease. Big data genomic assays have transformed our understanding of SLE by providing important insights into the molecular heterogeneity of this multigenic disease. Gene wide association studies have demonstrated more than 100 risk loci, supporting a model of multiple genetic hits increasing SLE risk in a non-linear fashion, and providing evidence of ancestral diversity in susceptibility loci. Epigenetic studies to determine the role of methylation, acetylation and non-coding RNAs have provided new understanding of the modulation of gene expression in SLE patients and identified new drug targets and biomarkers for SLE. Gene expression profiling has led to a greater understanding of the role of myeloid cells in the pathogenesis of SLE, confirmed roles for T and B cells in SLE, promoted clinical trials based on the prominent interferon signature found in SLE patients, and identified candidate biomarkers and cellular signatures to further drug development and drug repurposing. Gene expression studies are advancing our understanding of the underlying molecular heterogeneity in SLE and providing hope that patient stratification will expedite new therapies based on personal molecular signatures. Although big data analyses present unique interpretation challenges, both computationally and biologically, advances in machine learning applications may facilitate the ability to predict changes in SLE disease activity and optimize therapeutic strategies.
Collapse
|
268
|
Crow Y, Keshavan N, Barbet JP, Bercu G, Bondet V, Boussard C, Dedieu N, Duffy D, Hully M, Giardini A, Gitiaux C, Rice GI, Seabra L, Bader-Meunier B, Rahman S. Cardiac valve involvement in ADAR-related type I interferonopathy. J Med Genet 2019; 57:475-478. [DOI: 10.1136/jmedgenet-2019-106457] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Revised: 10/15/2019] [Accepted: 10/23/2019] [Indexed: 12/18/2022]
Abstract
BackgroundAdenosine deaminases acting on RNA (ADAR) mutations cause a spectrum of neurological phenotypes ranging from severe encephalopathy (Aicardi-Goutières syndrome) to isolated spastic paraplegia and are associated with enhanced type I interferon signalling. In children, non-neurological involvement in the type I interferonopathies includes autoimmune and rheumatological phenomena, with calcifying cardiac valve disease only previously reported in the context of MDA5 gain-of-function.ResultsWe describe three patients with biallelic ADAR mutations who developed calcifying cardiac valvular disease in late childhood (9.5–14 years). Echocardiography revealed progressive calcification of the valvular leaflets resulting in valvular stenosis and incompetence. Two patients became symptomatic with biventricular failure after 5–6.5 years. In one case, disease progressed to severe cardiac failure despite maximal medical management, with death occurring at 17 years. Another child received mechanical mitral and aortic valve replacement at 16 years with good postoperative outcome. Histological examination of the affected valves showed fibrosis and calcification.ConclusionsType I interferonopathies of differing genetic aetiology demonstrate an overlapping phenotypic spectrum which includes calcifying cardiac valvular disease. Individuals with ADAR-related type I interferonopathy may develop childhood-onset multivalvular stenosis and incompetence which can progress insidiously to symptomatic, and ultimately fatal, cardiac failure. Regular surveillance echocardiograms are recommended to detect valvular disease early.
Collapse
|
269
|
Tan RYY, Traylor M, Megy K, Duarte D, Deevi SVV, Shamardina O, Mapeta RP, Ouwehand WH, Gräf S, Downes K, Markus HS. How common are single gene mutations as a cause for lacunar stroke? A targeted gene panel study. Neurology 2019; 93:e2007-e2020. [PMID: 31719132 PMCID: PMC6913325 DOI: 10.1212/wnl.0000000000008544] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Accepted: 06/10/2019] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES To determine the frequency of rare and pertinent disease-causing variants in small vessel disease (SVD)-associated genes (such as NOTCH3, HTRA1, COL4A1, COL4A2, FOXC1, TREX1, and GLA) in cerebral SVD, we performed targeted gene sequencing in 950 patients with younger-onset apparently sporadic SVD stroke using a targeted sequencing panel. METHODS We designed a high-throughput sequencing panel to identify variants in 15 genes (7 known SVD genes, 8 SVD-related disorder genes). The panel was used to screen a population of 950 patients with younger-onset (≤70 years) MRI-confirmed SVD stroke, recruited from stroke centers across the United Kingdom. Variants were filtered according to their frequency in control databases, predicted effect, presence in curated variant lists, and combined annotation dependent depletion scores. Whole genome sequencing and genotyping were performed on a subset of patients to provide a direct comparison of techniques. The frequency of known disease-causing and pertinent variants of uncertain significance was calculated. RESULTS We identified previously reported variants in 14 patients (8 cysteine-changing NOTCH3 variants in 11 patients, 2 HTRA1 variants in 2 patients, and 1 missense COL4A1 variant in 1 patient). In addition, we identified 29 variants of uncertain significance in 32 patients. CONCLUSION Rare monogenic variants account for about 1.5% of younger onset lacunar stroke. Most are cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy variants, but the second most common gene affected is HTRA1. A high-throughput sequencing technology platform is an efficient, reliable method to screen for such mutations.
Collapse
Affiliation(s)
- Rhea Y Y Tan
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK.
| | - Matthew Traylor
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Karyn Megy
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Daniel Duarte
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Sri V V Deevi
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Olga Shamardina
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Rutendo P Mapeta
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Willem H Ouwehand
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Stefan Gräf
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Kate Downes
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| | - Hugh S Markus
- From the Stroke Research Group, Department of Clinical Neurosciences (R.Y.Y.T., M.T., H.S.M.), Department of Haematology (K.M., D.D., S.V.V.D., O.S., R.P.M., W.H.O., S.G., K.D.), and Division of Respiratory Medicine, Department of Medicine (S.G.), University of Cambridge; and NIHR BioResource: Rare Diseases (K.M., S.V.V.D., O.S., R.P.M., W.H.O., S.G., H.S.M.), Biomedical Campus, Cambridge, UK
| |
Collapse
|
270
|
Mammen AL, Allenbach Y, Stenzel W, Benveniste O. 239th ENMC International Workshop: Classification of dermatomyositis, Amsterdam, the Netherlands, 14-16 December 2018. Neuromuscul Disord 2019; 30:70-92. [PMID: 31791867 DOI: 10.1016/j.nmd.2019.10.005] [Citation(s) in RCA: 156] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Accepted: 10/21/2019] [Indexed: 12/29/2022]
Affiliation(s)
- Andrew L Mammen
- Muscle Disease Unit, Laboratory of Muscle Stem Cells and Gene Regulation, National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of Health, Bethesda, 50 South Drive, Building 50, Room 1146, MD 20892, United States.
| | - Yves Allenbach
- Department of Internal Medicine and Clinical Immunology, Pitié Salpetrière Hospital, AP-HP Sorbonne University, Paris, France
| | - Werner Stenzel
- Department of Neuropathology, Charité-Universitatsmedizin, Berlin, Germany
| | - Olivier Benveniste
- Department of Internal Medicine and Clinical Immunology, Pitié Salpetrière Hospital, AP-HP Sorbonne University, Paris, France
| | | |
Collapse
|
271
|
Mandhana R, Qian LK, Horvath CM. Constitutively Active MDA5 Proteins Are Inhibited by Paramyxovirus V Proteins. J Interferon Cytokine Res 2019; 38:319-332. [PMID: 30130154 DOI: 10.1089/jir.2018.0049] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Excessive interferon (IFN) production and signaling can lead to immunological and developmental defects giving rise to autoimmune diseases referred to collectively as "type I interferonopathies." A subset of these diseases is caused by monogenic mutations affecting proteins involved in nucleic acid sensing, homeostasis, and metabolism. Interferonopathic mutations in the cytosolic antiviral sensor MDA5 render it constitutively hyperactive, resulting in chronic IFN production and IFN-stimulated gene expression. Few therapeutic options are available for patients with interferonopathic diseases, but a large number of IFN evasion and antagonism strategies have evolved in viral pathogens that can counteract IFN production and signaling to enhance virus replication. To test the hypothesis that these natural IFN suppressors could be used to subdue the activity of interferonopathic signaling proteins, hyperactive MDA5 variants were assessed for susceptibility to a family of viral MDA5 inhibitors. In this study, Paramyxovirus V proteins were tested for their ability to counteract constitutively active MDA5 proteins. Results indicate that the V proteins are able to bind to and disrupt the signaling activity of these MDA5 proteins, irrespective of their specific mutations, reducing IFN production and IFN-stimulated gene expression to effectively suppress the hyperactive antiviral response.
Collapse
Affiliation(s)
- Roli Mandhana
- Department of Molecular Biosciences, Northwestern University , Evanston, Illinois
| | - Lily K Qian
- Department of Molecular Biosciences, Northwestern University , Evanston, Illinois
| | - Curt M Horvath
- Department of Molecular Biosciences, Northwestern University , Evanston, Illinois
| |
Collapse
|
272
|
West PK, Viengkhou B, Campbell IL, Hofer MJ. Microglia responses to interleukin-6 and type I interferons in neuroinflammatory disease. Glia 2019; 67:1821-1841. [PMID: 31033014 DOI: 10.1002/glia.23634] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 04/07/2019] [Accepted: 04/10/2019] [Indexed: 01/03/2025]
Abstract
Microglia are the resident macrophages of the central nervous system (CNS). They are a heterogenous, exquisitely responsive, and highly plastic cell population, which enables them to perform diverse roles. They sense and respond to the local production of many different signals, including an assorted range of cytokines. Microglia respond strongly to interleukin-6 (IL-6) and members of the type I interferon (IFN-I) family, IFN-alpha (IFN-α), and IFN-beta (IFN-β). Although these cytokines are essential in maintaining homeostasis and for activating and regulating immune responses, their chronic production has been linked to the development of distinct human neurological diseases, termed "cerebral cytokinopathies." IL-6 and IFN-α have been identified as key mediators in the pathogenesis of neuroinflammatory disorders including neuromyelitis optica and Aicardi-Goutières syndrome, respectively, whereas IFN-β has an emerging role as a causal factor in age-associated cognitive decline. One of the key features that unites these diseases is the presence of highly reactive microglia. The current understanding is that microglia contribute to the development of cerebral cytokinopathies and represent an important therapeutic target. However, it remains to be resolved whether microglia have beneficial or detrimental effects. Here we review and discuss what is currently known about the microglial response to IL-6 and IFN-I, based on both animal models and clinical studies. Foundational knowledge regarding the microglial response to IL-6 and IFN-I is now being used to devise therapeutic strategies to ameliorate neuroinflammation and promote repair: either through targeting microglia, or by targeting the reduction of CNS levels or downstream biological pathways of IL-6 or IFN-I.
Collapse
Affiliation(s)
- Phillip K West
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Barney Viengkhou
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Iain L Campbell
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| | - Markus J Hofer
- School of Life and Environmental Sciences, The Marie Bashir Institute for Infectious Diseases and Biosecurity, The Charles Perkins Centre, and The Bosch Institute, The University of Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
273
|
Sifuentes-Dominguez L, Starokadomskyy P, Welch J, Gurram B, Park JY, Koduru P, Burstein E. Mosaic Tetrasomy 9p Associated With Inflammatory Bowel Disease. J Crohns Colitis 2019; 13:1474-1478. [PMID: 31104071 PMCID: PMC6821155 DOI: 10.1093/ecco-jcc/jjz079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
The genetic basis of inflammatory bowel disease remains to be elucidated completely. Here we report on a patient with inflammatory bowel disease who has mosaic tetrasomy of the short arm of chromosome 9, a genomic region that harbours the type I interferon gene cluster. We show that increased interferon activation is present in peripheral blood and intestinal tissue from this patient, similar to previous reports of autoinflammatory organ damage driven by interferon activation in other patients with this chromosomal abnormality. To our knowledge, this is the first case of tetrasomy 9p-associated interferonopathy driving intestinal inflammation and highlights the role that type-I interferon pathways can play in the pathogenesis of intestinal inflammation.
Collapse
Affiliation(s)
- Luis Sifuentes-Dominguez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Corresponding author: Luis Sifuentes-Dominguez and Ezra Burstein, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Room J5.126, Dallas, TX 75390-9151, USA. Tel: 1-214-648-2008; Fax: 1-214-648-2022; ;
| | - Petro Starokadomskyy
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jacob Welch
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Bhaskar Gurram
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jason Y Park
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Eugene McDermott Center for Human Growth and Development, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Prasad Koduru
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ezra Burstein
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas, USA,Corresponding author: Luis Sifuentes-Dominguez and Ezra Burstein, Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Room J5.126, Dallas, TX 75390-9151, USA. Tel: 1-214-648-2008; Fax: 1-214-648-2022; ;
| |
Collapse
|
274
|
Briggs E, Crouch K, Lemgruber L, Hamilton G, Lapsley C, McCulloch R. Trypanosoma brucei ribonuclease H2A is an essential R-loop processing enzyme whose loss causes DNA damage during transcription initiation and antigenic variation. Nucleic Acids Res 2019; 47:9180-9197. [PMID: 31350892 PMCID: PMC6753483 DOI: 10.1093/nar/gkz644] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Revised: 07/09/2019] [Accepted: 07/16/2019] [Indexed: 12/21/2022] Open
Abstract
Ribonucleotides represent a threat to DNA genome stability and transmission. Two types of Ribonuclease H (RNase H) excise ribonucleotides when they form part of the DNA strand, or hydrolyse RNA when it base-pairs with DNA in structures termed R-loops. Loss of either RNase H is lethal in mammals, whereas yeast survives the absence of both enzymes. RNase H1 loss is tolerated by the parasite Trypanosoma brucei but no work has examined the function of RNase H2. Here we show that loss of T. brucei RNase H2 (TbRH2A) leads to growth and cell cycle arrest that is concomitant with accumulation of nuclear damage at sites of RNA polymerase (Pol) II transcription initiation, revealing a novel and critical role for RNase H2. Differential gene expression analysis reveals limited overall changes in RNA levels for RNA Pol II genes after TbRH2A loss, but increased perturbation of nucleotide metabolic genes. Finally, we show that TbRH2A loss causes R-loop and DNA damage accumulation in telomeric RNA Pol I transcription sites, also leading to altered gene expression. Thus, we demonstrate separation of function between two nuclear T. brucei RNase H enzymes during RNA Pol II transcription, but overlap in function during RNA Pol I-mediated gene expression during host immune evasion.
Collapse
Affiliation(s)
- Emma Briggs
- The Wellcome Centre for Integrative Parasitology, University of Glasgow, College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Kathryn Crouch
- The Wellcome Centre for Integrative Parasitology, University of Glasgow, College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Leandro Lemgruber
- The Wellcome Centre for Integrative Parasitology, University of Glasgow, College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Graham Hamilton
- Glasgow Polyomics, University of Glasgow, Wolfson Wohl Cancer Research Centre, Garscube Estate, Switchback Rd, Bearsden G61 1QH, UK
| | - Craig Lapsley
- The Wellcome Centre for Integrative Parasitology, University of Glasgow, College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| | - Richard McCulloch
- The Wellcome Centre for Integrative Parasitology, University of Glasgow, College of Medical, Veterinary and Life Sciences, Institute of Infection, Immunity and Inflammation, Sir Graeme Davies Building, 120 University Place, Glasgow G12 8TA, UK
| |
Collapse
|
275
|
Gatsiou A, Stellos K. Dawn of Epitranscriptomic Medicine. CIRCULATION-GENOMIC AND PRECISION MEDICINE 2019; 11:e001927. [PMID: 30354331 DOI: 10.1161/circgen.118.001927] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Medicine is at the crossroads of expanding disciplines. Prompt adaptation of medicine to each rapidly advancing research field, bridging bench to bedside, is a key step toward health improvement. Cardiovascular disease still ranks first among the mortality causes in the Western world, indicating a poor adaptation rate of cardiovascular medicine, albeit the gigantic scientific breakthroughs of this century. This urges the cardiovascular research field to explore novel concepts with promising prognostic and therapeutic potential. This review attempts to introduce the newly emerging field of epitranscriptome (or else known as RNA epigenetics) to cardiovascular researchers and clinicians summarizing its applications on health and disease. The traditionally perceived, intermediate carrier of genetic information or as contemporary revised as, occasionally, even the final product of gene expression, RNA, is dynamically subjected to >140 different kinds of chemical modifications determining its fate, which may profoundly impact the cellular responses and thus both health and disease course. Which are the most prevalent types of these RNA modifications, how are they catalyzed, how are they regulated, which role may they play in health and disease, and which are the implications for the cardiovascular medicine are few important questions that are discussed in the present review.
Collapse
Affiliation(s)
- Aikaterini Gatsiou
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany (A.G., K.S.).,Department of Cardiology, Center of Internal Medicine, Goethe University Frankfurt, Germany (A.G., K.S.).,German Center of Cardiovascular Research, Rhein-Main Partner Site, Frankfurt (A.G., K.S.)
| | - Konstantinos Stellos
- Institute of Cardiovascular Regeneration, Center of Molecular Medicine, Goethe University Frankfurt, Frankfurt am Main, Germany (A.G., K.S.).,Department of Cardiology, Center of Internal Medicine, Goethe University Frankfurt, Germany (A.G., K.S.).,German Center of Cardiovascular Research, Rhein-Main Partner Site, Frankfurt (A.G., K.S.).,Cardiovascular Research Centre, Institute of Genetic Medicine, Newcastle University, Newcastle upon Tyne, United Kingdom (K.S.).,Department of Cardiology, Freeman Hospital, Newcastle upon Tyne Hospitals National Health System Foundation Trust, United Kingdom (K.S.)
| |
Collapse
|
276
|
Morris ER, Taylor IA. The missing link: allostery and catalysis in the anti-viral protein SAMHD1. Biochem Soc Trans 2019; 47:1013-1027. [PMID: 31296733 PMCID: PMC7045340 DOI: 10.1042/bst20180348] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 06/04/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022]
Abstract
Vertebrate protein SAMHD1 (sterile-α-motif and HD domain containing protein 1) regulates the cellular dNTP (2'-deoxynucleoside-5'-triphosphate) pool by catalysing the hydrolysis of dNTP into 2'-deoxynucleoside and triphosphate products. As an important regulator of cell proliferation and a key player in dNTP homeostasis, mutations to SAMHD1 are implicated in hypermutated cancers, and germline mutations are associated with Chronic Lymphocytic Leukaemia and the inflammatory disorder Aicardi-Goutières Syndrome. By limiting the supply of dNTPs for viral DNA synthesis, SAMHD1 also restricts the replication of several retroviruses, such as HIV-1, and some DNA viruses in dendritic and myeloid lineage cells and resting T-cells. SAMHD1 activity is regulated throughout the cell cycle, both at the level of protein expression and post-translationally, through phosphorylation. In addition, allosteric regulation further fine-tunes the catalytic activity of SAMHD1, with a nucleotide-activated homotetramer as the catalytically active form of the protein. In cells, GTP and dATP are the likely physiological activators of two adjacent allosteric sites, AL1 (GTP) and AL2 (dATP), that bridge monomer-monomer interfaces to stabilise the protein homotetramer. This review summarises the extensive X-ray crystallographic, biophysical and molecular dynamics experiments that have elucidated important features of allosteric regulation in SAMHD1. We present a comprehensive mechanism detailing the structural and protein dynamics components of the allosteric coupling between nucleotide-induced tetramerization and the catalysis of dNTP hydrolysis by SAMHD1.
Collapse
Affiliation(s)
- Elizabeth R Morris
- Macromolecular Structure Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, U.K.
| | - Ian A Taylor
- Macromolecular Structure Laboratory, The Francis Crick Institute, 1 Midland Road, London NW1 1AT, U.K
| |
Collapse
|
277
|
Liu N, Chen J, Xu C, Shi T, Li J. Hereditary spastic paraplegia associated with a rare IFIH1 mutation: a case report and literature review. Hereditas 2019; 156:28. [PMID: 31427910 PMCID: PMC6693153 DOI: 10.1186/s41065-019-0104-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/30/2019] [Indexed: 11/29/2022] Open
Abstract
Here, the pathogenesis of an IFIH1 gene mutation is discussed through the analysis of a sporadic case of hereditary spastic paraplegia. Next-generation sequencing was performed for the patient and his family members to detect mutations at the IFIH1 locus. The patient and his father were found to carry the same heterozygous missense mutation (c.1093A > G; p.Gly495Arg), while the patient’s mother does not carry this mutation. This is the first report of this heterozygous IFIH1 mutation and it is predicted to be disease-causing.
Collapse
Affiliation(s)
- Nan Liu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province People's Republic of China
| | - Jiajun Chen
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province People's Republic of China
| | - Chuan Xu
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province People's Republic of China
| | - Tianji Shi
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province People's Republic of China
| | - Jia Li
- Department of Neurology, China-Japan Union Hospital of Jilin University, Changchun, Jilin Province People's Republic of China
| |
Collapse
|
278
|
Kumar G, Al Zoebie L, Duke C, Al Neyadi SM, Al Tenaiji A, ElGhazali G. Microcephaly and intracranial calcifications: Not always TORCH infection! J Paediatr Child Health 2019; 55:1000-1001. [PMID: 31386285 DOI: 10.1111/jpc.14535] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 11/27/2022]
Affiliation(s)
- Gurinder Kumar
- Department of Paediatrics, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Lama Al Zoebie
- Department of Paediatrics, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Christopher Duke
- Department of Cardiology, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Shaikha Mohamed Al Neyadi
- Department of Paediatric Intensive Care, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Amal Al Tenaiji
- Department of Paediatric Metabolic, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| | - Gehad ElGhazali
- Department of Immunology, Sheikh Khalifa Medical City, Abu Dhabi, United Arab Emirates
| |
Collapse
|
279
|
Saini AG, Gonda V, Bhatia V. Chronic Encephalopathy, Startle, and Intracranial Calcification: Think Beyond Intrauterine Infections. Pediatr Neurol 2019; 97:78-79. [PMID: 30922773 DOI: 10.1016/j.pediatrneurol.2019.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 01/17/2019] [Accepted: 01/23/2019] [Indexed: 11/19/2022]
Affiliation(s)
- Arushi Gahlot Saini
- Pediatric Neurology Unit, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India.
| | - Vijay Gonda
- Department of Pediatrics, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Vikas Bhatia
- Department of Radiodiagnosis, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| |
Collapse
|
280
|
RNASEH2B Related Adult-Onset Interferonopathy. J Clin Immunol 2019; 39:620-622. [DOI: 10.1007/s10875-019-00673-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2019] [Accepted: 07/17/2019] [Indexed: 11/30/2022]
|
281
|
Tirosh I, Spielman S, Barel O, Ram R, Stauber T, Paret G, Rubinsthein M, Pessach IM, Gerstein M, Anikster Y, Shukrun R, Dagan A, Adler K, Pode-Shakked B, Volkov A, Perelman M, Greenberger S, Somech R, Lahav E, Majmundar AJ, Padeh S, Hildebrandt F, Vivante A. Whole exome sequencing in childhood-onset lupus frequently detects single gene etiologies. Pediatr Rheumatol Online J 2019; 17:52. [PMID: 31362757 PMCID: PMC6668194 DOI: 10.1186/s12969-019-0349-y] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2019] [Accepted: 07/08/2019] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Systemic lupus erythematosus (SLE) comprise a diverse range of clinical manifestations. To date, more than 30 single gene causes of lupus/lupus like syndromes in humans have been identified. In the clinical setting, identifying the underlying molecular diagnosis is challenging due to phenotypic and genetic heterogeneity. METHODS We employed whole exome sequencing (WES) in patients presenting with childhood-onset lupus with severe and/or atypical presentations to identify cases that are explained by a single-gene (monogenic) cause. RESULTS From January 2015 to June 2018 15 new cases of childhood-onset SLE were diagnosed in Edmond and Lily Safra Children's Hospital. By WES we identified causative mutations in four subjects in five different genes: C1QC, SLC7A7, MAN2B1, PTEN and STAT1. No molecular diagnoses were established on clinical grounds prior to genetic testing. CONCLUSIONS We identified a significant fraction of monogenic SLE etiologies using WES and confirm the genetic locus heterogeneity in childhood-onset lupus. These results highlight the importance of establishing a genetic diagnosis for children with severe or atypical lupus by providing accurate and early etiology-based diagnoses and improving subsequent clinical management.
Collapse
Affiliation(s)
- Irit Tirosh
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0001 2107 2845grid.413795.dRheumatology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shiri Spielman
- 0000 0001 2107 2845grid.413795.dRheumatology Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Ortal Barel
- 0000 0001 2107 2845grid.413795.dThe Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Reut Ram
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel
| | - Tali Stauber
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics A Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Gideon Paret
- 0000 0001 2107 2845grid.413795.dIntensive care unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Marina Rubinsthein
- 0000 0001 2107 2845grid.413795.dIntensive care unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Itai M. Pessach
- 0000 0001 2107 2845grid.413795.dIntensive care unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Maya Gerstein
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Yair Anikster
- 0000 0001 2107 2845grid.413795.dMetabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Rachel Shukrun
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Adi Dagan
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Katerina Adler
- 0000 0001 2107 2845grid.413795.dThe Genomic Unit, Sheba Cancer Research Center, Sheba Medical Center, Tel Hashomer, Israel
| | - Ben Pode-Shakked
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0001 2107 2845grid.413795.dMetabolic Disease Unit, Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Alexander Volkov
- 0000 0001 2107 2845grid.413795.dPathology Department, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Marina Perelman
- 0000 0001 2107 2845grid.413795.dPathology Department, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Shoshana Greenberger
- 0000 0001 2107 2845grid.413795.dDepartment of Dermatology, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Raz Somech
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics A Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Einat Lahav
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics A Edmond and Lily Safra Children’s Hospital, Sheba Medical Center, Tel-Hashomer, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel ,0000 0001 2107 2845grid.413795.dNephrology Unit, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601 Ramat Gan, Israel
| | - Amar J. Majmundar
- 000000041936754Xgrid.38142.3cDivision of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Shai Padeh
- 0000 0001 2107 2845grid.413795.dDepartment of Pediatrics B, Edmond and Lily Safra Children’s Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601 Ramat Gan, Israel ,0000 0004 1937 0546grid.12136.37Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Friedhelm Hildebrandt
- 000000041936754Xgrid.38142.3cDivision of Nephrology, Department of Medicine, Boston Children’s Hospital, Harvard Medical School, Boston, MA USA
| | - Asaf Vivante
- Department of Pediatrics B, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel-Hashomer, 5265601, Ramat Gan, Israel. .,Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel. .,Nephrology Unit, Edmond and Lily Safra Children's Hospital, Sackler Faculty of Medicine, Sheba Medical Center, Tel Hashomer, 5265601, Ramat Gan, Israel.
| |
Collapse
|
282
|
Lamers MM, van den Hoogen BG, Haagmans BL. ADAR1: "Editor-in-Chief" of Cytoplasmic Innate Immunity. Front Immunol 2019; 10:1763. [PMID: 31404141 PMCID: PMC6669771 DOI: 10.3389/fimmu.2019.01763] [Citation(s) in RCA: 123] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2019] [Accepted: 07/11/2019] [Indexed: 12/12/2022] Open
Abstract
Specialized receptors that recognize molecular patterns such as double stranded RNA duplexes-indicative of viral replication-are potent triggers of the innate immune system. Although their activation is beneficial during viral infection, RNA transcribed from endogenous mobile genetic elements may also act as ligands potentially causing autoimmunity. Recent advances indicate that the adenosine deaminase ADAR1 through RNA editing is involved in dampening the canonical antiviral RIG-I-like receptor-, PKR-, and OAS-RNAse L pathways to prevent autoimmunity. However, this inhibitory effect must be overcome during viral infections. In this review we discuss ADAR1's critical role in balancing immune activation and self-tolerance.
Collapse
|
283
|
Nezos A, Makri P, Gandolfo S, De Vita S, Voulgarelis M, Crow MK, Mavragani CP. TREX1 variants in Sjogren's syndrome related lymphomagenesis. Cytokine 2019; 132:154781. [PMID: 31326279 DOI: 10.1016/j.cyto.2019.154781] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2018] [Revised: 06/28/2019] [Accepted: 07/09/2019] [Indexed: 12/28/2022]
Abstract
Genetic variants of the three-prime repair exonuclease 1 (TREX1) -an exonuclease involved in DNA repair and degradation- have been previously found to increase susceptibility to Aicardi Goutieres syndrome, familial chilblain lupus and systemic lupus erythematosus. We aimed to explore whether TREX1 common variants could influence the risk of primary Sjogren's syndrome (SS) and SS-related lymphoma. Three single nucleotide polymorphisms (SNPs) of the TREX1 gene (rs11797, rs3135941 and rs3135945) were evaluated in 229 SS, 89 SS-lymphoma (70 SS-MALT and 19 SS non-MALT) and 240 healthy controls by PCR-based assays. In available 52 peripheral blood and 26 minor salivary gland tissues from our SS cohort, mRNA expression of type I interferon (IFN) related genes and TREX1 was determined by real-time PCR. Significantly decreased prevalence of rs11797 A minor allele was detected in SS patients complicated by non-MALT lymphoma compared to controls (ΟR [95% CI]: 0.4 [0.2-0.9], p-value: 0.02). SS patients carrying the rs11797 AA genotype had increased type I IFN related gene mRNA expression in minor salivary gland tissues. These data support genetically related dampened type I IFN production as an additional mechanism for SS-related lymphomagenesis.
Collapse
Affiliation(s)
- Adrianos Nezos
- Department of Physiology, School of Medicine, National University of Athens, Athens, Greece
| | - Panagiota Makri
- Department of Physiology, School of Medicine, National University of Athens, Athens, Greece
| | - Saviana Gandolfo
- Rheumatology Clinic, Department of Medical and Biological Sciences, Azienda Ospedaliero-Universitaria 'S. Maria della Misericordia', Udine, Italy
| | - Salvatore De Vita
- Rheumatology Clinic, Department of Medical and Biological Sciences, Azienda Ospedaliero-Universitaria 'S. Maria della Misericordia', Udine, Italy
| | - Michael Voulgarelis
- Department of Pathophysiology, School of Medicine, National University of Athens, Athens, Greece
| | - Mary K Crow
- Mary Kirkland Center for Lupus Research, Hospital for Special Surgery, Weill Medical College of Cornell University, New York, NY, USA
| | - Clio P Mavragani
- Department of Physiology, School of Medicine, National University of Athens, Athens, Greece; Department of Pathophysiology, School of Medicine, National University of Athens, Athens, Greece; Joint Academic Rheumatology Program, National and Kapodistrian University of Athens, School of Medicine, Athens, Greece.
| |
Collapse
|
284
|
Williams JS, Lujan SA, Zhou ZX, Burkholder AB, Clark AB, Fargo DC, Kunkel TA. Genome-wide mutagenesis resulting from topoisomerase 1-processing of unrepaired ribonucleotides in DNA. DNA Repair (Amst) 2019; 84:102641. [PMID: 31311768 DOI: 10.1016/j.dnarep.2019.102641] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/28/2019] [Accepted: 07/02/2019] [Indexed: 01/10/2023]
Abstract
Ribonucleotides are the most common non-canonical nucleotides incorporated into DNA during replication, and their processing leads to mutations and genome instability. Yeast mutation reporter systems demonstrate that 2-5 base pair deletions (Δ2-5bp) in repetitive DNA are a signature of unrepaired ribonucleotides, and that these events are initiated by topoisomerase 1 (Top1) cleavage. However, a detailed understanding of the frequency and locations of ribonucleotide-dependent mutational events across the genome has been lacking. Here we present the results of genome-wide mutational analysis of yeast strains deficient in Ribonucleotide Excision Repair (RER). We identified mutations that accumulated over thousands of generations in strains expressing either wild-type or variant replicase alleles (M644G Pol ε, L612M Pol δ, L868M Pol α) that confer increased ribonucleotide incorporation into DNA. Using a custom-designed mutation-calling pipeline called muver (for mutationes verificatae), we observe a number of surprising mutagenic features. This includes a 24-fold preferential elevation of AG and AC relative to AT dinucleotide deletions in the absence of RER, suggesting specificity for Top1-initiated deletion mutagenesis. Moreover, deletion rates in di- and trinucleotide repeat tracts increase exponentially with tract length. Consistent with biochemical and reporter gene mutational analysis, these deletions are no longer observed upon deletion of TOP1. Taken together, results from these analyses demonstrate the global impact of genomic ribonucleotide processing by Top1 on genome integrity.
Collapse
Affiliation(s)
- Jessica S Williams
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Scott A Lujan
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Zhi-Xiong Zhou
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Adam B Burkholder
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Alan B Clark
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - David C Fargo
- Integrative Bioinformatics Support Group, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA
| | - Thomas A Kunkel
- Genome Integrity and Structural Biology Laboratory, National Institute of Environmental Health Sciences, US National Institutes of Health, Department of Health and Human Services, Research Triangle Park, NC, USA.
| |
Collapse
|
285
|
Affiliation(s)
- Tetsuji Sawada
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Daiki Fujimori
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| | - Yusuke Yamamoto
- Department of Rheumatology, Tokyo Medical University Hospital, Tokyo, Japan
| |
Collapse
|
286
|
García Pérez JL, Alarcón-Riquelme ME. The TREX1 Dinosaur Bites the Brain through the LINE. Cell Stem Cell 2019; 21:287-288. [PMID: 28886359 DOI: 10.1016/j.stem.2017.08.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
In this issue of Cell Stem Cell, Thomas et al. (2017) define the nature of accumulated ssDNA present in the neuron and astrocyte cytoplasm of TREX1 mutated stem cell-derived organoids. Accumulated ssDNAs are derived from LINE-1 endogenous retroelements, providing new clues as to the development of Aicardi-Goutières syndrome in the neural system.
Collapse
Affiliation(s)
- José Luis García Pérez
- Pfizer - University of Granada - Andalusian Government Center for Genomics and Oncological Research (GENYO), PTS, 18016, Granada, Spain; Medical Research Council Human Genetics Unit, Institute of Genetics and Molecular Medicine, University of Edinburgh, Western General Hospital, Edinburgh EH4 2XU, UK
| | - Marta E Alarcón-Riquelme
- Pfizer - University of Granada - Andalusian Government Center for Genomics and Oncological Research (GENYO), PTS, 18016, Granada, Spain; Unit for Chronic Inflammatory Diseases, Institute for Environmental Medicine, Karolinska Institute, Solna, 171 77, Sweden.
| |
Collapse
|
287
|
Durcan L, O'Dwyer T, Petri M. Management strategies and future directions for systemic lupus erythematosus in adults. Lancet 2019; 393:2332-2343. [PMID: 31180030 DOI: 10.1016/s0140-6736(19)30237-5] [Citation(s) in RCA: 357] [Impact Index Per Article: 59.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2017] [Revised: 01/03/2019] [Accepted: 01/23/2019] [Indexed: 12/13/2022]
Abstract
Systemic lupus erythematosus (SLE) is an autoimmune disease characterised by the loss of self-tolerance and formation of nuclear autoantigens and immune complexes resulting in inflammation of multiple organs. The clinical presentation of SLE is heterogeneous, can involve one or more organs, including the skin, kidneys, joints, and nervous system, and take a chronic or relapsing and remitting disease course. SLE is most common in women and in those of non-white ethnicity. Because of the multitude of presentations, manifestations, and serological abnormalities in patients with SLE, diagnosis can be challenging. Therapeutic approaches predominantly involve immunomodulation and immunosuppression and are targeted to the specific organ manifestation, with the aim of achieving low disease activity. Despite many treatment advances and improved diagnostics, SLE continues to cause substantial morbidity and premature mortality. Current management strategies, although helpful, are limited by high failure rates and toxicity. An overreliance on corticosteroid therapy contributes to much of the long-term organ damage. In this Seminar, we outline the classification criteria for SLE, current treatment strategies and medications, the evidence supporting their use, and explore potential future therapies.
Collapse
Affiliation(s)
- Laura Durcan
- Department of Rheumatology, Beaumont Hospital, Dublin, Ireland; Department of Medicine, The Royal College of Surgeons of Ireland, Dublin, Ireland.
| | - Tom O'Dwyer
- School of Physiotherapy, Trinity College, Dublin, Ireland
| | - Michelle Petri
- Division of Rheumatology, Johns Hopkins University School of Medicine, Baltimore, MA, USA
| |
Collapse
|
288
|
Abstract
Human brain organoids, generated from pluripotent stem cells, have emerged as a promising technique for modeling early stages of human neurodevelopment in controlled laboratory conditions. Although the applications for disease modeling in a dish have become routine, the use of these brain organoids as evolutionary tools is only now getting momentum. Here, we will review the current state of the art on the use of brain organoids from different species and the molecular and cellular insights generated from these studies. Besides, we will discuss how this model might be beneficial for human health and the limitations and future perspectives of this technology.
Collapse
Affiliation(s)
- Alysson R. Muotri
- Department of Pediatrics/Rady Children's Hospital San Diego, University of California San Diego, School of Medicine, La Jolla, CA, USA
- Department of Cellular & Molecular Medicine, University of California San Diego, School of Medicine, La Jolla, CA, USA
- UCSD Stem Cell Programme, University of California San Diego, School of Medicine, La Jolla, CA, USA
- Center for Academic Research and Training in Anthropogeny (CARTA), La Jolla, CA, USA
- Kavli Institute for Brain and Mind, University of California San Diego, School of Medicine, La Jolla, CA, USA
| |
Collapse
|
289
|
Garau J, Cavallera V, Valente M, Tonduti D, Sproviero D, Zucca S, Battaglia D, Battini R, Bertini E, Cappanera S, Chiapparini L, Crasà C, Crichiutti G, Dalla Giustina E, D'Arrigo S, De Giorgis V, De Simone M, Galli J, La Piana R, Messana T, Moroni I, Nardocci N, Panteghini C, Parazzini C, Pichiecchio A, Pini A, Ricci F, Saletti V, Salvatici E, Santorelli FM, Sartori S, Tinelli F, Uggetti C, Veneselli E, Zorzi G, Garavaglia B, Fazzi E, Orcesi S, Cereda C. Molecular Genetics and Interferon Signature in the Italian Aicardi Goutières Syndrome Cohort: Report of 12 New Cases and Literature Review. J Clin Med 2019; 8:jcm8050750. [PMID: 31130681 PMCID: PMC6572054 DOI: 10.3390/jcm8050750] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Revised: 05/04/2019] [Accepted: 05/08/2019] [Indexed: 01/30/2023] Open
Abstract
Aicardi-Goutières syndrome (AGS) is a genetically determined early onset encephalopathy characterized by cerebral calcification, leukodystrophy, and increased expression of interferon-stimulated genes (ISGs). Up to now, seven genes (TREX1, RNASEH2B, RNASEH2C, RNASEH2A, ADAR1, SAMHD1, IFIH1) have been associated with an AGS phenotype. Next Generation Sequencing (NGS) analysis was performed on 51 AGS patients and interferon signature (IS) was investigated in 18 AGS patients and 31 healthy controls. NGS identified mutations in 48 of 51 subjects, with three patients demonstrating a typical AGS phenotype but not carrying mutations in known AGS-related genes. Five mutations, in RNASEH2B, SAMHD1 and IFIH1 gene, were not previously reported. Eleven patients were positive and seven negatives for the upregulation of interferon signaling (IS > 2.216). This work presents, for the first time, the genetic data of an Italian cohort of AGS patients, with a higher percentage of mutations in RNASEH2B and a lower frequency of mutations in TREX1 than those seen in international series. RNASEH2B mutated patients showed a prevalence of negative IS consistent with data reported in the literature. We also identified five novel pathogenic mutations that warrant further functional investigation. Exome/genome sequencing will be performed in future studies in patients without a mutation in AGS-related genes.
Collapse
Affiliation(s)
- Jessica Garau
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy.
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Vanessa Cavallera
- Unit of Child and Adolescence Neurology, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Marialuisa Valente
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Davide Tonduti
- Pediatric Neurology Unit, V. Buzzi Children's Hospital, 20154 Milan, Italy.
| | - Daisy Sproviero
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Susanna Zucca
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Domenica Battaglia
- Child Neuropsichiatry, Department of Woman and Child Health and Public Health, Fondazione Policlinico Universitario A. Gemelli IRCCS, Università Cattolica del Sacro Cuore, 00168 Roma, Italy.
| | - Roberta Battini
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy.
| | - Enrico Bertini
- Unit of Neuromuscular and Neurodegenerative Disorders, Bambino Gesù Children's Hospital, IRCCS, 00165 Rome, Italy.
| | - Silvia Cappanera
- S.O.D. Neuropsichiatria Infantile, Ospedali Riuniti "G. Salesi", 60123 Ancona, Italy.
| | - Luisa Chiapparini
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Camilla Crasà
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | | | - Elvio Dalla Giustina
- Child Neurology Unit, IRCCS, Santa Maria Nuova Hospital, 42123 Reggio Emilia, Italy.
| | - Stefano D'Arrigo
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Valentina De Giorgis
- Unit of Child and Adolescence Neurology, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Micaela De Simone
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, 25123 Brescia, Italy.
| | - Jessica Galli
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, 25123 Brescia, Italy.
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy.
| | - Roberta La Piana
- Department of Neuroradiology andLaboratory of Neurogenetics of Motion, Montreal Neurological Institute and Hospital, McGill University, Montreal, QC H3A2B4, Canada.
| | - Tullio Messana
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, 40139 Bologna, Italy.
| | - Isabella Moroni
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Nardo Nardocci
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Celeste Panteghini
- Medical Genetics and Neurogenetics Unit, Movement Disorders Diagnostic Section, Fondazione Irccs IstitutoNeurologico Carlo Besta, 20133 Milan, Italy.
| | - Cecilia Parazzini
- Department of Pediatric Radiology and Neuroradiology, V. Buzzi Children's Hospital, 20154 Milan, Italy.
| | - Anna Pichiecchio
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy.
- Neuroradiology Unit, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Antonella Pini
- Child Neurology Unit, IRCCS Istituto delle Scienze Neurologiche, 40139 Bologna, Italy.
| | - Federica Ricci
- Unit of Child Neurology and Psychiatry, University Hospital Città della Salute e della Scienza, 10126 Turin, Italy.
| | - Veronica Saletti
- Developmental Neurology Division, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Elisabetta Salvatici
- Clinical Department of Pediatrics San Paolo Hospital - ASST Santi Paolo Carlo, 20142 Milano, Italy.
| | | | - Stefano Sartori
- Paediatric Neurology and Neurophysiology Unit, Department of Women's and Children's Health, University Hospital of Padua, 35128 Padua, Italy.
| | - Francesca Tinelli
- Department of Developmental Neuroscience, IRCCS Stella Maris Foundation, 56128 Pisa, Italy.
| | - Carla Uggetti
- Neuroradiology Unit, Department of Radiology, ASST Santi Paolo e Carlo, San Carlo Borromeo Hospital, 20153 Milan, Italy.
| | - Edvige Veneselli
- Child Neuropsychiatry Unit, IRCCS Giannina Gaslini Institute DINOGMI, University of Genoa, 16147 Genoa, Italy.
| | - Giovanna Zorzi
- Department of Pediatric Neuroscience, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133 Milan, Italy.
| | - Barbara Garavaglia
- Medical Genetics and Neurogenetics Unit, Movement Disorders Diagnostic Section, Fondazione Irccs IstitutoNeurologico Carlo Besta, 20133 Milan, Italy.
| | - Elisa Fazzi
- Child Neurology and Psychiatry Unit, ASST Spedali Civili of Brescia, 25123 Brescia, Italy.
- Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy.
| | - Simona Orcesi
- Department of Brain and Behavioral Sciences, University of Pavia, 27100 Pavia, Italy.
- Unit of Child and Adolescence Neurology, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| | - Cristina Cereda
- Genomic and Post-Genomic Center, IRCCS Mondino Foundation, 27100 Pavia, Italy.
| |
Collapse
|
290
|
Wanrooij PH, Chabes A. Ribonucleotides in mitochondrial DNA. FEBS Lett 2019; 593:1554-1565. [PMID: 31093968 DOI: 10.1002/1873-3468.13440] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2019] [Revised: 05/09/2019] [Accepted: 05/09/2019] [Indexed: 01/05/2023]
Abstract
The incorporation of ribonucleotides (rNMPs) into DNA during genome replication has gained substantial attention in recent years and has been shown to be a significant source of genomic instability. Studies in yeast and mammals have shown that the two genomes, the nuclear DNA (nDNA) and the mitochondrial DNA (mtDNA), differ with regard to their rNMP content. This is largely due to differences in rNMP repair - whereas rNMPs are efficiently removed from the nuclear genome, mitochondria lack robust mechanisms for removal of single rNMPs incorporated during DNA replication. In this minireview, we describe the processes that determine the frequency of rNMPs in the mitochondrial genome and summarise recent findings regarding the effect of incorporated rNMPs on mtDNA stability and function.
Collapse
Affiliation(s)
- Paulina H Wanrooij
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden
| | - Andrei Chabes
- Department of Medical Biochemistry and Biophysics, Umeå University, Sweden.,Laboratory for Molecular Infection Medicine Sweden, Umeå University, Sweden
| |
Collapse
|
291
|
Eyk CLV, Samaraweera SE, Scott A, Webber DL, Harvey DP, Mecinger O, O’Keefe LV, Cropley JE, Young P, Ho J, Suter C, Richards RI. Non-self mutation: double-stranded RNA elicits antiviral pathogenic response in a Drosophila model of expanded CAG repeat neurodegenerative diseases. Hum Mol Genet 2019; 28:3000-3012. [DOI: 10.1093/hmg/ddz096] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/11/2019] [Accepted: 05/06/2019] [Indexed: 12/16/2022] Open
Abstract
Abstract
Inflammation is activated prior to symptoms in neurodegenerative diseases, providing a plausible pathogenic mechanism. Indeed, genetic and pharmacological ablation studies in animal models of several neurodegenerative diseases demonstrate that inflammation is required for pathology. However, while there is growing evidence that inflammation-mediated pathology may be the common mechanism underlying neurodegenerative diseases, including those due to dominantly inherited expanded repeats, the proximal causal agent is unknown. Expanded CAG.CUG repeat double-stranded RNA causes inflammation-mediated pathology when expressed in Drosophila. Repeat dsRNA is recognized by Dicer-2 as a foreign or ‘non-self’ molecule triggering both antiviral RNA and RNAi pathways. Neither of the RNAi pathway cofactors R2D2 nor loquacious are necessary, indicating antiviral RNA activation. RNA modification enables avoidance of recognition as ‘non-self’ by the innate inflammatory surveillance system. Human ADAR1 edits RNA conferring ‘self’ status and when co-expressed with expanded CAG.CUG dsRNA in Drosophila the pathology is lost. Cricket Paralysis Virus protein CrPV-1A is a known antagonist of Argonaute-2 in Drosophila antiviral defense. CrPV-1A co-expression also rescues pathogenesis, confirming anti-viral-RNA response. Repeat expansion mutation therefore confers ‘non-self’ recognition of endogenous RNA, thereby providing a proximal, autoinflammatory trigger for expanded repeat neurodegenerative diseases.
Collapse
Affiliation(s)
- Clare L van Eyk
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Saumya E Samaraweera
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Andrew Scott
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Dani L Webber
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - David P Harvey
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Olivia Mecinger
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Louise V O’Keefe
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Jennifer E Cropley
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Paul Young
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Joshua Ho
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
- School of Biomedical Sciences, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Pokfulam, Hong Kong
| | - Catherine Suter
- Victor Chang Cardiac Research Institute, Lowy Packer Building, Liverpool St, Darlinghurst, Sydney 2010, Australia
- Faculty of Medicine, University of New South Wales, Kensington, New South Wales 2042, Australia
| | - Robert I Richards
- Department of Molecular and Biomedical Science, School of Biological Sciences, The University of Adelaide, Adelaide, South Australia 5000, Australia
| |
Collapse
|
292
|
Mustelin T, Lood C, Giltiay NV. Sources of Pathogenic Nucleic Acids in Systemic Lupus Erythematosus. Front Immunol 2019; 10:1028. [PMID: 31139185 PMCID: PMC6519310 DOI: 10.3389/fimmu.2019.01028] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 04/23/2019] [Indexed: 12/19/2022] Open
Abstract
A hallmark of systemic lupus erythematosus (SLE), and several related autoimmune diseases, is the presence of autoantibodies against nucleic acids and nucleic acid-binding proteins, as well as elevated type I interferons (IFNs), which appear to be instrumental in disease pathogenesis. Here we discuss the sources and proposed mechanisms by which a range of cellular RNA and DNA species can become pathogenic and trigger the nucleic acid sensors that drive type I interferon production. Potentially SLE-promoting DNA may originate from pieces of chromatin, from mitochondria, or from reverse-transcribed cellular RNA, while pathogenic RNA may arise from mis-localized, mis-processed, ancient retroviral, or transposable element-derived transcripts. These nucleic acids may leak out from dying cells to be internalized and reacted to by immune cells or they may be generated and remain to be sensed intracellularly in immune or non-immune cells. The presence of aberrant DNA or RNA is normally counteracted by effective counter-mechanisms, the loss of which result in a serious type I IFN-driven disease called Aicardi-Goutières Syndrome. However, in SLE it remains unclear which mechanisms are most critical in precipitating disease: aberrant RNA or DNA, overly sensitive sensor mechanisms, or faulty counter-acting defenses. We propose that the clinical heterogeneity of SLE may be reflected, in part, by heterogeneity in which pathogenic nucleic acid molecules are present and which sensors and pathways they trigger in individual patients. Elucidation of these events may result in the recognition of distinct "endotypes" of SLE, each with its distinct therapeutic choices.
Collapse
Affiliation(s)
- Tomas Mustelin
- Division of Rheumatology, Department of Medicine, University of Washington, Seattle, WA, United States
| | | | | |
Collapse
|
293
|
Abstract
PURPOSE OF REVIEW Autoimmune diseases are of unknown origin, and they represent significant causes of morbidity and mortality. Here, we review new developments in the understanding of their pathogenesis that have led to development of well tolerated and effective treatments. RECENT FINDINGS In addition to the long-recognized genetic impact of the HLA locus, interferon regulatory factors, PTPN22, STAT4, and NOX have been implicated in pathogenesis of systemic lupus erythematosus (SLE) and rheumatoid arthritis (RA). Smoking, ultraviolet light, diet, and microbiota exert strong environmental influence on development of RA and SLE. Metabolism has been recognized as a critical integrator of genetic and environmental factors, and it controls immune cell differentiation both under physiological and pathological conditions. SUMMARY With the advent of high-throughput genetic, proteomic, and metabolomic technologies, the field of medicine has been shifting towards systems-based and personalized approaches to diagnose and treat common conditions, including rheumatic diseases. Regulatory checkpoints of metabolism and signal transduction, such as glucose utilization, mitochondrial electron transport, JAK, mTOR, and AMPK pathway activation, and production of pro-inflammatory cytokines IL-1, IL-6, and IL-17 have presented new targets for therapeutic intervention. This review amalgamates recent discoveries in genetics and metabolomics with immunological pathways of pathogenesis in rheumatic diseases.
Collapse
Affiliation(s)
- Eric Liu
- Division of Rheumatology, Departments of Medicine, Microbiology and Immunology, Biochemistry and Molecular Biology, State University of New York, Upstate Medical University, College of Medicine, Syracuse, New York, USA
| | | |
Collapse
|
294
|
Deasy SK, Uehara R, Vodnala SK, Yang HH, Dass RA, Hu Y, Lee MP, Crouch RJ, Hunter KW. Aicardi-Goutières syndrome gene Rnaseh2c is a metastasis susceptibility gene in breast cancer. PLoS Genet 2019; 15:e1008020. [PMID: 31125342 PMCID: PMC6553800 DOI: 10.1371/journal.pgen.1008020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2019] [Revised: 06/06/2019] [Accepted: 04/26/2019] [Indexed: 12/14/2022] Open
Abstract
Breast cancer is the second leading cause of cancer-related deaths in the United States, with the majority of these deaths due to metastatic lesions rather than the primary tumor. Thus, a better understanding of the etiology of metastatic disease is crucial for improving survival. Using a haplotype mapping strategy in mouse and shRNA-mediated gene knockdown, we identified Rnaseh2c, a scaffolding protein of the heterotrimeric RNase H2 endoribonuclease complex, as a novel metastasis susceptibility factor. We found that the role of Rnaseh2c in metastatic disease is independent of RNase H2 enzymatic activity, and immunophenotyping and RNA-sequencing analysis revealed engagement of the T cell-mediated adaptive immune response. Furthermore, the cGAS-Sting pathway was not activated in the metastatic cancer cells used in this study, suggesting that the mechanism of immune response in breast cancer is different from the mechanism proposed for Aicardi-Goutières Syndrome, a rare interferonopathy caused by RNase H2 mutation. These results suggest an important novel, non-enzymatic role for RNASEH2C during breast cancer progression and add Rnaseh2c to a panel of genes we have identified that together could determine patients with high risk for metastasis. These results also highlight a potential new target for combination with immunotherapies and may contribute to a better understanding of the etiology of Aicardi-Goutières Syndrome autoimmunity.
Collapse
Affiliation(s)
- Sarah K. Deasy
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
- Institute for Biomedical Sciences, The George Washington University, Washington, District of Columbia, United States of America
| | - Ryo Uehara
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Suman K. Vodnala
- Surgery Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Howard H. Yang
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Randall A. Dass
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Ying Hu
- Center for Biomedical Informatics and Information Technology, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Maxwell P. Lee
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Robert J. Crouch
- SFR, Division of Intramural Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kent W. Hunter
- Laboratory of Cancer Biology and Genetics, National Cancer Institute, National Institutes of Health, Bethesda, Maryland, United States of America
| |
Collapse
|
295
|
Amari S, Tsukamoto K, Ishiguro A, Yanagi K, Kaname T, Ito Y. An extremely severe case of Aicardi-Goutières syndrome 7 with a novel variant in IFIH1. Eur J Med Genet 2019; 63:103646. [PMID: 30965144 DOI: 10.1016/j.ejmg.2019.04.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/25/2019] [Accepted: 04/02/2019] [Indexed: 12/17/2022]
Abstract
We describe herein an extremely severe case of Aicardi-Goutières syndrome 7 (AGS7). The female patient was the daughter of nonconsanguineous parents and developed cardiomegaly, pericardial effusion, splenomegaly, and intracranial calcification during the fetal period. Because her cardiotocogram showed a non-reassuring fetal status, she was delivered at 29 weeks and 4 days of gestation by an emergency cesarean section. After birth, she suffered from respiratory distress, pulmonary hypertension, refractory fever, recurrent thrombocytopenia, and abdominal distention caused by hepatomegaly and ascites. She showed a lenticulostriate vasculopathy, which was compatible with the fetal intracranial calcification. Despite various intensive care procedures, she died of gradually progressive pulmonary hypertension at 3 months of age. After her death, whole exome sequencing on the patient and the parents was performed and revealed a novel, de novo, heterozygous mutation in the IFIH1 gene (IFIH1:NM_022168:exon12:c.2439A > T:p.Glu813Asp). On the basis of the mutation and the clinical features, the diagnosis was AGS7. Although AGS7 has been regarded as a relatively mild subtype of Aicardi-Goutières syndrome, this case indicates that the c.2439A > T variant of AGS7 can be fatal in early infancy.
Collapse
Affiliation(s)
- Shoichiro Amari
- Division of Neonatology, Center for Maternal-Fetal, Neonatal, and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan.
| | - Keiko Tsukamoto
- Division of Neonatology, Center for Maternal-Fetal, Neonatal, and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Akira Ishiguro
- Division of Hematology, National Center for Child Health and Development, Tokyo, Japan
| | - Kumiko Yanagi
- Department of Genome Medicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Tadashi Kaname
- Department of Genome Medicine, National Research Institute for Child Health and Development, Tokyo, Japan
| | - Yushi Ito
- Division of Neonatology, Center for Maternal-Fetal, Neonatal, and Reproductive Medicine, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
296
|
Potenski CJ, Epshtein A, Bianco C, Klein HL. Genome instability consequences of RNase H2 Aicardi-Goutières syndrome alleles. DNA Repair (Amst) 2019; 84:102614. [PMID: 30975634 DOI: 10.1016/j.dnarep.2019.04.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 01/07/2023]
Abstract
The RNase H2 complex is a conserved heterotrimeric enzyme that degrades RNA:DNA hybrids and promotes excision of rNMPs misincorporated during DNA replication. Failure to remove ribonucleotides from DNA leads to genomic instability in yeast and humans. The monogenic Aicardi-Goutières syndrome (AGS) results from mutation in one of several genes, among which are those encoding the RNase H2 subunits. The complete cellular and genomic consequences of RNASEH2 mutations and the precise connection to disease remain unclear. To learn more about the effect of RNASEH2 mutations on the cell, we used yeast as a model of AGS disease. We have generated yeast strains bearing AGS-associated mutations in RNASEH2 genes. There is a range of disease presentation in patients bearing these RNASEH2 variants. Here we report on in vivo phenotypes of genomic instability, including mutation and recombination rates, and synthetic gene interactions. These phenotypes provide insight into molecular consequences of RNASEH2 mutations, and lay the groundwork for further study of genomic instability as a contributing factor to AGS disease.
Collapse
Affiliation(s)
- Catherine J Potenski
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Anastasiya Epshtein
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA
| | - Christopher Bianco
- Department of Microbiology, New York University School of Medicine, New York, NY 10016, USA
| | - Hannah L Klein
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, NY 10016, USA.
| |
Collapse
|
297
|
Tonduti D, Izzo G, D'Arrigo S, Riva D, Moroni I, Zorzi G, Cavallera V, Pichiecchio A, Uggetti C, Veggiotti P, Orcesi S, Chiapparini L, Parazzini C. Spontaneous MRI improvement and absence of cerebral calcification in Aicardi-Goutières syndrome: Diagnostic and disease-monitoring implications. Mol Genet Metab 2019; 126:489-494. [PMID: 30826161 DOI: 10.1016/j.ymgme.2019.02.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/20/2018] [Revised: 02/20/2019] [Accepted: 02/22/2019] [Indexed: 10/27/2022]
Abstract
BACKGROUND Aicardi-Goutières syndrome (AGS) is a rare genetic leukoencephalopathy related to inappropriate activation of type I interferon. Neuroradiological findings are typically characterized by white matter abnormalities, cerebral atrophy and cerebral calcification. The disease usually manifests itself during the first year of life in the form of an initial "encephalitic-like" phase followed by a chronic phase of stabilization of the neurological signs. Recently new therapeutic strategies have been proposed aimed at blocking the abnormal activation of the interferon cascade. MATERIALS AND METHODS We reviewed clinical and MRI findings in three young RNASEH2B-mutated patients studied with serial CT and MRI studies. RESULTS All three patients presented clinical and MRI features consistent with AGS but, very unexpectedly, an improving neuroradiological course. In patient 1, the MRI improvement was noted some months after treatment with high-dose steroid and IVIg treatment; in patients 2 and 3 it occurred spontaneously. Patient 2 did not show cerebral calcification on CT images. CONCLUSIONS Our series highlights the possibility of spontaneous neuroradiological improvement in AGS2 patients, as well as the possibility of absence of cerebral calcification in AGS. The study underlines the need for extreme caution when using MRI as an outcome measure in therapeutic trials specific for this disease. MRI follow-up studies in larger series are necessary to describe the natural course of AGS.
Collapse
Affiliation(s)
- Davide Tonduti
- Pediatric Neurology Unit, V. Buzzi Children's Hospital, Milan, Italy.
| | - Giana Izzo
- Department of Pediatric Radiology and Neuroradiology, V. Buzzi Children's Hospital, Milan, Italy
| | - Stefano D'Arrigo
- Child Neurology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Daria Riva
- Child Neurology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Isabella Moroni
- Child Neurology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Giovanna Zorzi
- Child Neurology Department, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Vanessa Cavallera
- Child and Adolescent Neurology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Anna Pichiecchio
- Neuroradiology Unit, IRCCS Mondino Foundation, Pavia, Italy; Department of Brain and Behavioural Sciences, University of Pavia, Pavia, Italy
| | - Carla Uggetti
- Neuroradiology Unit, Department of Radiology, ASST Santi Paolo e Carlo, San Carlo Borromeo Hospital, Milan, Italy
| | - Pierangelo Veggiotti
- Pediatric Neurology Unit, V. Buzzi Children's Hospital, Milan, Italy; Biomedical and Clinical Science Department, University of Milan, Milan, Italy
| | - Simona Orcesi
- Child and Adolescent Neurology Department, IRCCS Mondino Foundation, Pavia, Italy
| | - Luisa Chiapparini
- Neuroradiology Unit, Fondazione IRCCS Istituto Neurologico Carlo Besta, Milan, Italy
| | - Cecilia Parazzini
- Department of Pediatric Radiology and Neuroradiology, V. Buzzi Children's Hospital, Milan, Italy
| |
Collapse
|
298
|
Mohammad SS, Paget SP, Dale RC. Current therapies and therapeutic decision making for childhood-onset movement disorders. Mov Disord 2019; 34:637-656. [PMID: 30919519 DOI: 10.1002/mds.27661] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Accepted: 02/12/2019] [Indexed: 12/13/2022] Open
Abstract
Movement disorders differ in children to adults. First, neurodevelopmental movement disorders such as tics and stereotypies are more prevalent than parkinsonism, and second, there is a genomic revolution which is now explaining many early-onset dystonic syndromes. We outline an approach to children with movement disorders starting with defining the movement phenomenology, determining the level of functional impairment due to abnormal movements, and screening for comorbid psychiatric conditions and cognitive impairments which often contribute more to disability than the movements themselves. The rapid improvement in our understanding of the etiology of movement disorders has resulted in an increasing focus on precision medicine, targeting treatable conditions and defining modifiable disease processes. We profile some of the key disease-modifying therapies in metabolic, neurotransmitter, inflammatory, and autoimmune conditions and the increasing focus on gene or cellular therapies. When no disease-modifying therapies are possible, symptomatic therapies are often all that is available. These classically target dopaminergic, cholinergic, alpha-adrenergic, or GABAergic neurochemistry. Increasing interest in neuromodulation has highlighted that some clinical syndromes respond better to DBS, and further highlights the importance of "disease-specific" therapies with a future focus on individualized therapies according to the genomic findings or disease pathways that are disrupted. We summarize some pragmatic applications of symptomatic therapies, neuromodulation techniques, and some rehabilitative interventions and provide a contemporary overview of treatment in childhood-onset movement disorders. © 2019 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Shekeeb S Mohammad
- Kids Neuroscience Centre, The Kids Research Institute at the Children's Hospital at Westmead, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia.,Movement Disorders Unit, T.Y. Nelson Department of Neurology, the Children's Hospital at Westmead and Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Simon P Paget
- Kids Rehab, the Children's Hospital at Westmead and Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| | - Russell C Dale
- Kids Neuroscience Centre, The Kids Research Institute at the Children's Hospital at Westmead, Brain and Mind Centre, Faculty of Medicine and Health, University of Sydney, Westmead, NSW, Australia.,Movement Disorders Unit, T.Y. Nelson Department of Neurology, the Children's Hospital at Westmead and Sydney Medical School, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
299
|
|
300
|
Brain organoids as a model system for human neurodevelopment and disease. Semin Cell Dev Biol 2019; 95:93-97. [PMID: 30904636 DOI: 10.1016/j.semcdb.2019.03.002] [Citation(s) in RCA: 39] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Revised: 02/26/2019] [Accepted: 03/19/2019] [Indexed: 12/15/2022]
Abstract
The ability to reproduce early stages of human neurodevelopment in the laboratory is one of the most exciting fields in modern neuroscience. The inaccessibility of the healthy human brain developing in utero has delayed our understanding of the initial steps in the formation of one of the most complex tissues in the body. Animal models, postmortem human tissues and cellular systems have been instrumental in contributing to our understanding of the human brain. However, all model systems have intrinsic limitations. The emerging field of brain organoids, which are three-dimensional self-assembled multicellular structures derived from human pluripotent stem cells, offers a promising complementary cellular model for the study of the human brain. Here, we will discuss the initial experiments that were the foundation for this emerging field, highlight recent uses of the technology and offer our perspective on future directions that might guide further exploratory experimentation to improve the human brain organoid model system.
Collapse
|