1
|
Lundqvist T, Stenlid R, Halldin M. Hormonal Crossroads in Inborn Errors of the Metabolism Impact of Puberty and Dietary Interventions on Metabolic Health. Metabolites 2025; 15:235. [PMID: 40278364 PMCID: PMC12029320 DOI: 10.3390/metabo15040235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2025] [Revised: 03/21/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
Background/Objectives: Inborn errors of metabolism (IEMs) represent a diverse group of genetic disorders characterized by enzymatic defects that disrupt metabolic pathways, leading to toxic metabolite accumulation, deficits, or impaired macromolecule synthesis. While strict dietary interventions are critical for managing many of these conditions, hormonal and metabolic changes during puberty introduce new challenges. Advancements in early diagnosis and treatment have significantly extended the lifespan of individuals with IEMs. However, this increased longevity is associated with heightened risks of new medical problems, including obesity, insulin resistance, and type 2 diabetes mellitus (T2DM), as these complications share mechanistic features with those seen in obesity and T2DM. Methods: This mini-review examines current knowledge of the intricate interplay between pubertal hormones and metabolic pathways in IEM patients. Results: We address critical questions, such as if puberty intensifies the risk of metabolic derangements in these individuals and if there is a metabolic intersection where these disorders converge, leading to shared complications. We highlight the impact of puberty-induced hormonal fluctuations, such as growth hormone (GH) surges and sex steroid activity, on disorders like phenylketonuria, urea cycle defects, and fatty acid oxidation disorders. Moreover, we explore the role of dietary interventions in mitigating or exacerbating these effects, emphasizing the importance of balancing nutritional needs during growth spurts. Conclusions: A multidisciplinary approach integrating endocrinology, nutrition, and emerging therapies is advocated to optimize metabolic health during puberty. Addressing these challenges is critical for improving long-term outcomes for individuals with IEMs, particularly during this pivotal developmental phase.
Collapse
Affiliation(s)
- Thomas Lundqvist
- Department of Women’s and Children’s Health, Karolinska Institutet, 171 76 Stockholm, Sweden
- Unit for Pediatric Endocrinology and Metabolic Disorders, Karolinska University Hospital, 171 76 Stockholm, Sweden
| | - Rasmus Stenlid
- Department of Medical Cell Biology, Uppsala University, 751 23 Uppsala, Sweden
| | - Maria Halldin
- Department of Women’s and Children’s Health, Karolinska Institutet, 171 76 Stockholm, Sweden
- Unit for Pediatric Endocrinology and Metabolic Disorders, Karolinska University Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
2
|
Wang J, Ding X, Jia K, Chen H, An G, Zhao Q, Shen D, Qiu Z, Zhang X, Qian H, Xia D. BmWARS inhibits BmNPV infection via the PI3K-Akt pathway. BULLETIN OF ENTOMOLOGICAL RESEARCH 2025:1-14. [PMID: 40125613 DOI: 10.1017/s000748532500015x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
Bombyx mori Tryptophanyl-tRNA synthetase (BmWARS) belongs to the family of Ic-like aminoacyl-tRNA synthetases (aaRSs), whose specific recognition of the substrate Trp, tRNA, maintains the fidelity of protein synthesis. In this study, BmWARS was cloned and characterized from the midgut of the silkworm, Bombyx mori, resulting in an open reading frame (ORF) with a full length of 1,149 bp, which can encode 382 Aa. BmWARS is localized in the cytoplasm, and is expressed in all tissues of the silkworm, with higher expression in the testis, ovary, silk gland and malpighian tubule. The expression of BmWARS was significantly up-regulated in the midgut and silk gland after infection with Bombyx mori nuclear polyhedrosis virus (BmNPV). In addition, overexpression of BmWARS inhibited BmNPV infection and replication extremely significantly, while interference with BmWARS expression promoted BmNPV infection and replication. Analysis of the immune pathways in which BmWARS may be involved revealed that the expression of the key genes of the PI3K-Akt pathway, BmPI3K, BmAkt, BmPDK1, BmeIF4E, BmS6, and p-Akt protein was significantly reduced, whereas the expression of BmPTEN, BmFoxO, and BmCaspase9 was significantly increased in the cells that overexpressed BmWARS and were infected with BmNPV. Meanwhile, the results of the study interfering with the expression of BmWARS were completely opposite to those of the study overexpressing BmWARS. This is the first report that BmWARS has antiviral effects in Bombyx mori. Moreover, BmWARS inhibits BmNPV infection and replication in Bombyx mori cells by promoting apoptosis and inhibiting cell proliferation.
Collapse
Affiliation(s)
- Jinyang Wang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Xiangrui Ding
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Kaifang Jia
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Haiyu Chen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Guorong An
- Yancheng Agricultural College, Yancheng College of Agricultural Science and Technology Vocational, Yancheng, China
| | - Qiaoling Zhao
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Dongxu Shen
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Zhiyong Qiu
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Xuelian Zhang
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Heying Qian
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| | - Dingguo Xia
- Jiangsu Key Laboratory of Sericultural and Animal Biotechnology, School of Biotechnology, Jiangsu University of Science and Technology, Zhenjiang, China
- Key Laboratory of Silkworm and Mulberry Genetic Improvement, Ministry of Agriculture and Rural Affairs, Sericultural Scientific Research Center, Chinese Academy of Agricultural Sciences, Zhenjiang, China
| |
Collapse
|
3
|
Hoytema van Konijnenburg EMM, Rohof J, Kok G, van Hasselt PM, van Karnebeek CD, Muffels IJJ, Fuchs SA. Setting the Stage for Treatment of Aminoacyl-tRNA Synthetase (ARS)1-Deficiencies: Phenotypic Characterization and a Review of Treatment Effects. J Inherit Metab Dis 2025; 48:e70017. [PMID: 40044141 PMCID: PMC11882346 DOI: 10.1002/jimd.70017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/09/2025]
Abstract
Aminoacyl-transfer RNA (tRNA) synthetases (ARSs) are key enzymes for protein translation. The number of identified patients with recessive ARS1 deficiencies is rapidly increasing. Initially, only supportive care was available, but in recent years beneficial effects of targeted amino acid supplementation have been described. To allow early treatment and prevention of symptoms, rapid recognition is necessary, as well as insight into the natural history to evaluate treatment effects. We performed a scoping literature search for clinical characteristics and treatment effects of patients with ARS1 deficiencies. Symptoms were matched to Human Phenotype Ontology terms. We identified 438 patients with 20 different ARS1 deficiencies. Overall mortality was 22%. Neurological symptoms were most prevalent across all ARS1 deficiencies (in 87% of patients), including neurodevelopmental disorder (79%), microcephaly (50%) and seizures (46%). Growth issues and ophthalmological symptoms were also prevalent in many ARS1 deficiencies. Two distinct phenotypical clusters were seen: one with multisystemic disease including liver- and lung disease and another with a predominantly neurological phenotype. Supplementation with cognate amino acids was described in 21 patients, with beneficial effects (e.g., improvements in growth, development, liver and lung disease) in the majority. Treatment did not alleviate the most severe phenotypes. Specific symptoms relate to (a cluster of) specific ARS1 deficiencies; the mechanism is not yet understood. Multi-organ involvement should trigger inclusion of ARS1 genes in the diagnostic work-up. Treatment with cognate amino acids is promising, but it remains challenging to distinguish treatment effects from natural history. Synopsis: Treatment with cognate amino acids in ARS1 deficiencies is promising, but it remains challenging to distinguish treatment effects from natural history.
Collapse
Affiliation(s)
- Eva M. M. Hoytema van Konijnenburg
- Department of Metabolic DiseasesWilhelmina Children's Hospital University Medical Centre Utrechtthe Netherlands
- On Behalf of United for Metabolic DiseasesAmsterdamthe Netherlands
| | - Joline Rohof
- Department of Metabolic DiseasesWilhelmina Children's Hospital University Medical Centre Utrechtthe Netherlands
| | - Gautam Kok
- Department of Metabolic DiseasesWilhelmina Children's Hospital University Medical Centre Utrechtthe Netherlands
- On Behalf of United for Metabolic DiseasesAmsterdamthe Netherlands
| | - Peter M. van Hasselt
- Department of Metabolic DiseasesWilhelmina Children's Hospital University Medical Centre Utrechtthe Netherlands
- On Behalf of United for Metabolic DiseasesAmsterdamthe Netherlands
| | - Clara D. van Karnebeek
- On Behalf of United for Metabolic DiseasesAmsterdamthe Netherlands
- Emma Center for Personalized Medicine, Department of Pediatrics and Human GeneticsAmsterdam UMCthe Netherlands
| | - Irena J. J. Muffels
- Department of Metabolic DiseasesWilhelmina Children's Hospital University Medical Centre Utrechtthe Netherlands
- On Behalf of United for Metabolic DiseasesAmsterdamthe Netherlands
| | - Sabine A. Fuchs
- Department of Metabolic DiseasesWilhelmina Children's Hospital University Medical Centre Utrechtthe Netherlands
- On Behalf of United for Metabolic DiseasesAmsterdamthe Netherlands
| |
Collapse
|
4
|
Del Greco C, Kuo ME, Smith DEC, Mendes MI, Salamons GS, Nemcovic M, Kodrikova R, Sestak S, Stancheva M, Antonellis A. Loss-of-Function CARS1 Variants in a Patient With Microcephaly, Developmental Delay, and a Brittle Hair Phenotype. Mol Genet Genomic Med 2025; 13:e70078. [PMID: 39963003 PMCID: PMC11833167 DOI: 10.1002/mgg3.70078] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Revised: 01/17/2025] [Accepted: 02/04/2025] [Indexed: 02/21/2025] Open
Abstract
BACKGROUND Mutations in cysteinyl-tRNA synthetase (CARS1) have been implicated in a multisystem disease including microcephaly, developmental delay, and brittle hair and nail phenotypes. METHODS Here, we present a patient with hepatopathy, hypothyroidism, short stature, developmental delay, microcephaly, muscular hypotonia, brittle hair, and ataxia. The patient underwent exome sequencing to identify potentially pathogenic genetic variants. In addition, identified variants were assessed using yeast complementation assays to determine functional consequences. RESULTS Exome sequencing determined that the patient is compound heterozygous for p.Arg341His and p.Arg370Trp CARS1. Yeast complementation assays showed that the p.Arg341His variant has a hypomorphic effect and that the p.Arg370Trp variant causes a complete loss-of-function effect. CONCLUSION This study is the second report of pathogenic CARS1 variants and expands the allelic and phenotypic heterogeneity of CARS1-associated disease.
Collapse
Affiliation(s)
- Christina Del Greco
- Department of Human GeneticsUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Molly E. Kuo
- Department of Human GeneticsUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Cellular and Molecular Biology ProgramUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Medical Scientist Training ProgramUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| | - Desiree E. C. Smith
- Laboratory Genetic Metabolic Diseases, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Marisa I. Mendes
- Laboratory Genetic Metabolic Diseases, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Gajja S. Salamons
- Laboratory Genetic Metabolic Diseases, Amsterdam UMCUniversity of AmsterdamAmsterdamthe Netherlands
| | - Marek Nemcovic
- Institute of Chemistry, Department of GlycobiologySlovak Academy of SciencesBratislavaSlovakia
| | - Rebeka Kodrikova
- Institute of Chemistry, Department of GlycobiologySlovak Academy of SciencesBratislavaSlovakia
| | - Sergej Sestak
- Institute of Chemistry, Department of GlycobiologySlovak Academy of SciencesBratislavaSlovakia
| | | | - Anthony Antonellis
- Department of Human GeneticsUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Cellular and Molecular Biology ProgramUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
- Department of NeurologyUniversity of Michigan Medical SchoolAnn ArborMichiganUSA
| |
Collapse
|
5
|
Wu J, Fan Y, Huo F, Deng J, Wang Q, Shen Y. Case report: Infantile pulmonary alveolar proteinosis associated with cytosolic isoleucyl-tRNA synthetase deficiency. Front Pharmacol 2025; 16:1487993. [PMID: 39950113 PMCID: PMC11821940 DOI: 10.3389/fphar.2025.1487993] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/10/2025] [Indexed: 02/16/2025] Open
Abstract
Cytosolic isoleucyl-tRNA synthetase (IARS1) deficiency, an exceptionally rare autosomal recessive inherited disorder, is characterized by multiple system involvement, including growth retardation, intellectual developmental disorder, hypotonia, and hepatopathy. Pulmonary alveolar proteinosis (PAP) is a rare phenotype of IARS1 deficiency, having been reported in only two siblings from the same family. In this study, we present a case of IARS1 deficiency in a 5-month-old boy, who exhibited PAP as the initial and predominant manifestation. Additionally, whole-exome sequencing identified compound heterozygous variants in the IARS1 gene (c.2428C>T/c.128T>C), both of which are novel observations.
Collapse
Affiliation(s)
- Jie Wu
- Emergency Department, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Yimu Fan
- Emergency Department, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Feng Huo
- Emergency Department, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Jie Deng
- Neurology Department, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Quan Wang
- Pediatric Intensive Care Unit, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
| | - Yuelin Shen
- Respiratory Department II, National Clinical Research Center for Respiratory Diseases, Beijing Children’s Hospital, Capital Medical University, National Center for Children’s Health, Beijing, China
- Respiratory Department, Children’s Hospital Affiliated to Zhengzhou University, Henan Children’s hospital, Zhengzhou Children’s Hospital, Zhengzhou, China
| |
Collapse
|
6
|
Kok G, Schene IF, Ilcken EF, Alcaraz PS, Mendes M, Smith DEC, Salomons G, Shehata S, Jans JJM, Maroofian R, Hoek TA, van Es RM, Rehmann H, Nieuwenhuis ES, Vos HR, Fuchs SA. Isoleucine-to-valine substitutions support cellular physiology during isoleucine deprivation. Nucleic Acids Res 2025; 53:gkae1184. [PMID: 39657787 PMCID: PMC11724295 DOI: 10.1093/nar/gkae1184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 11/08/2024] [Accepted: 11/13/2024] [Indexed: 12/12/2024] Open
Abstract
Aminoacyl-tRNA synthetases (ARSs) couple tRNAs with their corresponding amino acids. While ARSs can bind structurally similar amino acids, extreme specificity is ensured by subsequent editing activity. Yet, we found that upon isoleucine (I) restriction, healthy fibroblasts consistently incorporated valine (V) into proteins at isoleucine codons, resulting from misacylation of tRNAIle with valine by wildtype IARS1. Using a dual-fluorescent reporter of translation, we found that valine supplementation could fully compensate for isoleucine depletion and restore translation to normal levels in healthy, but not IARS1 deficient cells. Similarly, the antiproliferative effects of isoleucine deprivation could be fully restored by valine supplementation in healthy, but not IARS1 deficient cells. This indicates I > V substitutions help prevent translational termination and maintain cellular function in human primary cells during isoleucine deprivation. We suggest that this is an example of a more general mechanism in mammalian cells to preserve translational speed at the cost of translational fidelity in response to (local) amino acid deficiencies.
Collapse
Affiliation(s)
- Gautam Kok
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Imre F Schene
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Eveline F Ilcken
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Paula Sobrevals Alcaraz
- Center for Molecular Medicine, University Medical Center Utrecht, Oncode Institute, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Marisa I Mendes
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Desiree E C Smith
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Gajja Salomons
- Laboratory Genetic Metabolic Diseases, Amsterdam University Medical Centers, Meibergdreef 9, 1105 AZ Amsterdam, The Netherlands
| | - Sawsan Shehata
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Judith J M Jans
- Laboratory of Metabolic Diseases, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Reza Maroofian
- Department of Neuromuscular Disorders, UCL Queen Square Institute of Neurology, Queen Square, London WC1N 3BG, United Kingdom
| | - Tim A Hoek
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, Uppsalalaan 8, 3584 CT Utrecht, The Netherlands
| | - Robert M van Es
- Center for Molecular Medicine, University Medical Center Utrecht, Oncode Institute, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Holger Rehmann
- Department Energy and Biotechnology, Flensburg University of Applied Sciences, Kanzleistraße 91–93 24943 Flensburg, Germany
| | - Edward E S Nieuwenhuis
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| | - Harmjan R Vos
- Center for Molecular Medicine, University Medical Center Utrecht, Oncode Institute, Heidelberglaan 100, 3584 CX Utrecht, The Netherlands
| | - Sabine A Fuchs
- Department of Metabolic Diseases, Wilhelmina Children’s Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA Utrecht, The Netherlands
| |
Collapse
|
7
|
Giunta-Stibb H, Hackett B. Interstitial lung disease in the newborn. J Perinatol 2025; 45:13-23. [PMID: 38956315 DOI: 10.1038/s41372-024-02036-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 05/30/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024]
Abstract
Although relatively rare, interstitial lung diseases may present with respiratory distress in the newborn period. Most commonly these include developmental and growth disorders, disorders of surfactant synthesis and homeostasis, pulmonary interstitial glycogenosis, and neuroendocrine cell hyperplasia of infancy. Although the diagnosis of these disorders is sometimes made based on clinical presentation and imaging, due to the significant overlap between disorders and phenotypic variability, lung biopsy or, increasingly genetic testing is needed for diagnosis. These diseases may result in significant morbidity and mortality. Effective medical treatment options are in some cases limited and/or invasive. The genetic basis for some of these disorders has been identified, and with increased utilization of exome and whole genome sequencing even before lung biopsy, further insights into their genetic etiologies should become available.
Collapse
Affiliation(s)
- Hannah Giunta-Stibb
- Divisions of Neonatology and Pulmonology, Department of Pediatrics, University of Rochester Medical Center, 601 Elmwood Ave, Rochester, NY, 14642, USA.
| | - Brian Hackett
- Mildred Stahlman Division of Neonatology, Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| |
Collapse
|
8
|
Samuels TN, Wu F, Mahmood M, Abuzaid WA, Sun N, Moresco A, Siu VM, O'Donoghue P, Heinemann IU. Transfer RNA and small molecule therapeutics for aminoacyl-tRNA synthetase diseases. FEBS J 2024. [PMID: 39702998 DOI: 10.1111/febs.17361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/08/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
Aminoacyl-tRNA synthetases catalyze the ligation of a specific amino acid to its cognate tRNA. The resulting aminoacyl-tRNAs are indispensable intermediates in protein biosynthesis, facilitating the precise decoding of the genetic code. Pathogenic alleles in the aminoacyl-tRNA synthetases can lead to several dominant and recessive disorders. To date, disease-specific treatments for these conditions are largely unavailable. We review pathogenic human synthetase alleles, the molecular and cellular mechanisms of tRNA synthetase diseases, and emerging approaches to allele-specific treatments, including small molecules and nucleic acid-based therapeutics. Current treatment approaches to rescue defective or dysfunctional tRNA synthetase mutants include supplementation with cognate amino acids and delivery of cognate tRNAs to alleviate bottlenecks in translation. Complementary approaches use inhibitors to target the integrated stress response, which can be dysregulated in tRNA synthetase diseases.
Collapse
Affiliation(s)
- Tristan N Samuels
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Fanqi Wu
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Maria Mahmood
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Wajd A Abuzaid
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Nancy Sun
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Canada
| | - Angelica Moresco
- Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Children's Health Research Institute, London, Canada
| | - Victoria M Siu
- Department of Paediatrics, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Children's Health Research Institute, London, Canada
| | - Patrick O'Donoghue
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Department of Chemistry, Western University, London, Canada
| | - Ilka U Heinemann
- Department of Biochemistry, Schulich School of Medicine and Dentistry, Western University, London, Canada
- Children's Health Research Institute, London, Canada
| |
Collapse
|
9
|
Uehara T, Seki E, Nonoda Y, Kumaki T, Tsuyusaki Y, Aida N, Enomoto Y, Ishikura K, Kurosawa K. Two siblings with acute necrotizing encephalopathy associated with variants of LARS1. Am J Med Genet A 2024; 194:e63803. [PMID: 38923116 DOI: 10.1002/ajmg.a.63803] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Revised: 05/18/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024]
Abstract
Acute necrotizing encephalopathy (ANE) is a rapidly progressive encephalopathy of unknown etiology. The underlying mechanisms are highly heterogeneous, often including genetic backgrounds. Variants of LARS1, encoding the leucyl-tRNA synthetase 1, are responsible for infantile liver failure syndrome 1. We describe two siblings with ANE caused by compound heterozygous variants of LARS1. Patient 1 was a 17-month-old girl. She presented with generalized seizure and liver dysfunction due to influenza type A infection. Brain magnetic resonance imaging on day 4 of onset showed diffuse high-intensity signals consistent with ANE. She died on day 10. Patient 2, a younger male sibling of patient 1, had mild to moderate developmental delay and growth failure at the age of 18 months. He showed a markedly elevated level of transaminases triggered by infection with human herpesvirus 6. On day 4 of onset, he had generalized seizures. Brain computed tomography showed a diffuse symmetrical hypodensity consistent with ANE. He died on day 7. Whole exome sequencing identified the compound heterozygous variants in LARS1 (NM_020117.11) as c.83_88delinsAATGGGATA, p.(Arg28_Phe30delinsLysTryAspIle) and c.1283C>T, p.(Pro428Leu) in both siblings. The severe neurologic phenotype, found in our patients, reflects the complicated pathogenesis of LARS1-related disorder.
Collapse
Affiliation(s)
- Takeshi Uehara
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Eijun Seki
- Department of Neurology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yutaka Nonoda
- Department of Pediatrics, Kitasato University School of Medicine, Sagamihara, Japan
| | - Tatsuro Kumaki
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yu Tsuyusaki
- Department of Neurology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Noriko Aida
- Department of Radiology, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Yumi Enomoto
- Clinical Research Institute, Kanagawa Children's Medical Center, Yokohama, Japan
| | - Kenji Ishikura
- Department of Pediatrics, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kenji Kurosawa
- Division of Medical Genetics, Kanagawa Children's Medical Center, Yokohama, Japan
| |
Collapse
|
10
|
Borie R, Berteloot L, Kannengiesser C, Griese M, Cazes A, Crestani B, Hadchouel A, Debray MP. Rare genetic interstitial lung diseases: a pictorial essay. Eur Respir Rev 2024; 33:240101. [PMID: 39537246 PMCID: PMC11558537 DOI: 10.1183/16000617.0101-2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Accepted: 08/22/2024] [Indexed: 11/16/2024] Open
Abstract
The main monogenic causes of pulmonary fibrosis in adults are mutations in telomere-related genes. These mutations may be associated with extrapulmonary signs (hepatic, haematological and dermatological) and typically present radiologically as usual interstitial pneumonia or unclassifiable fibrosis. In children, the monogenic causes of pulmonary fibrosis are dominated by mutations in surfactant-related genes. These mutations are not associated with extrapulmonary signs and often manifest radiologically as unclassifiable fibrosis with cysts that can lead to chest wall deformities in adults. This review discusses these mutations, along with most of the monogenic causes of interstitial lung disease, including interferon-related genes, mutations in genes causing cystic lung disease, Hermansky-Pudlak syndrome, pulmonary alveolar proteinosis, lysinuric protein intolerance and lysosomal storage disorders, and their pulmonary and extrapulmonary manifestations.
Collapse
Affiliation(s)
- Raphael Borie
- Université Paris Cité, Inserm, PHERE, Paris, France
- Hôpital Bichat, APHP, Service de Pneumologie A, Centre constitutif du centre de référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | - Laureline Berteloot
- Service d'Imagerie Pédiatrique, Hôpital universitaire Necker-Enfants malades, Paris, France
- INSERM U1163, Paris, France
| | | | - Matthias Griese
- Department of Pediatric Pneumology, Dr von Hauner Children's Hospital, Ludwig-Maximilians-University, German Center for Lung Research, Munich, Germany
| | - Aurelie Cazes
- Département d'Anatomo-Pathologie, Hôpital Bichat, AP-HP, Paris, France
| | - Bruno Crestani
- Université Paris Cité, Inserm, PHERE, Paris, France
- Hôpital Bichat, APHP, Service de Pneumologie A, Centre constitutif du centre de référence des Maladies Pulmonaires Rares, FHU APOLLO, Paris, France
| | - Alice Hadchouel
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France
- INSERM U1151, Institut Necker Enfants Malades, Université Paris Cité, Paris, France
| | - Marie Pierre Debray
- Service de Radiologie, Hopital Bichat, APHP, Université Paris Cité, Paris, France
| |
Collapse
|
11
|
Pelayo G, Paiva Coelho M, Correia J, Bandeira A, Nogueira C, Vilarinho L, Martins E. Phenotyping mitochondrial glutamyl-tRNA synthetase deficiency (EARS2): A case series and systematic literature review. Neurobiol Dis 2024; 200:106644. [PMID: 39173847 DOI: 10.1016/j.nbd.2024.106644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 07/26/2024] [Accepted: 08/18/2024] [Indexed: 08/24/2024] Open
Abstract
Mitochondrial glutamyl-aminoacyl tRNA synthetase deficiency, stemming from biallelic mutations in the EARS2 gene, was first described in 2012. With <50 cases reported globally, this condition exhibits a distinct phenotype of neonatal or childhood-onset, often referred to as leukoencephalopathy with thalamus and brainstem involvement and high lactate (LTBL). It has also been one of the few reversible mitochondrial disorders described. The natural history of these patients is poorly documented, ranging from clinical and radiological improvement to early death. Herein, we detail three cases from our centre, including follow-up on the Portuguese patient reported by Steenweg et al., These cases illustrate the phenotypic spectrum: i) rapidly progressive neonatal presentation with lactic acidemia and corpus callosum agenesis, leading to early death; ii) early onset with a severe, slowly progressive course; iii) early onset with a milder phenotype, showing some improvement and mild neurological symptoms. Additionally, we conducted a systematic literature review on cases of EARS2-deficient patients, focusing on clinical manifestations, laboratory findings, radiological aspects, and disease progression over time, along with respective data analysis. "Patients with EARS2 deficiency typically present within the first year of life with a well-defined neurometabolic disorder picture, often including hypotonia and/or spasticity, along with neurodevelopmental delay or regression. There are no pathognomonic features specific to EARS2 deficiency, and no genotype-phenotype correlation has been identified." Comparing to initial characterization by Steenweg et al., this analysis reveals an expanded disease spectrum. We propose a novel strategy for clustering phenotypes into severe, moderate, or mild disease based on initial presentation, seemingly correlating with disease progression. The paucity of data on the disease's natural history highlights the need for a multicentric approach to enhance understanding and management. TAKE-HOME MESSAGE: Analysis of all cases published with EARS2 deficiency allows for establish disease spectrum and a novel strategy for clustering phenotypes which correlate to disease progression.
Collapse
Affiliation(s)
- Gonçalo Pelayo
- Reference Centre for Metabolic Disorders, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Margarida Paiva Coelho
- Reference Centre for Metabolic Disorders, Centro Hospitalar Universitário de Santo António, Porto, Portugal.
| | - Joana Correia
- Reference Centre for Metabolic Disorders, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Anabela Bandeira
- Reference Centre for Metabolic Disorders, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| | - Célia Nogueira
- Newborn Screening, Metabolism and Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisboa, Portugal
| | - Laura Vilarinho
- Newborn Screening, Metabolism and Genetics Unit, Human Genetics Department, National Institute of Health Doutor Ricardo Jorge, Lisboa, Portugal
| | - Esmeralda Martins
- Reference Centre for Metabolic Disorders, Centro Hospitalar Universitário de Santo António, Porto, Portugal
| |
Collapse
|
12
|
Meyer-Schuman R, Cale AR, Pierluissi JA, Jonatzke KE, Park YN, Lenk GM, Oprescu SN, Grachtchouk MA, Dlugosz AA, Beg AA, Meisler MH, Antonellis A. A model organism pipeline provides insight into the clinical heterogeneity of TARS1 loss-of-function variants. HGG ADVANCES 2024; 5:100324. [PMID: 38956874 PMCID: PMC11284558 DOI: 10.1016/j.xhgg.2024.100324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 06/27/2024] [Accepted: 06/27/2024] [Indexed: 07/04/2024] Open
Abstract
Aminoacyl-tRNA synthetases (ARSs) are ubiquitously expressed, essential enzymes that complete the first step of protein translation: ligation of amino acids to cognate tRNAs. Genes encoding ARSs have been implicated in myriad dominant and recessive phenotypes, the latter often affecting multiple tissues but with frequent involvement of the central and peripheral nervous systems, liver, and lungs. Threonyl-tRNA synthetase (TARS1) encodes the enzyme that ligates threonine to tRNATHR in the cytoplasm. To date, TARS1 variants have been implicated in a recessive brittle hair phenotype. To better understand TARS1-related recessive phenotypes, we engineered three TARS1 missense variants at conserved residues and studied these variants in Saccharomyces cerevisiae and Caenorhabditis elegans models. This revealed two loss-of-function variants, including one hypomorphic allele (R433H). We next used R433H to study the effects of partial loss of TARS1 function in a compound heterozygous mouse model (R432H/null). This model presents with phenotypes reminiscent of patients with TARS1 variants and with distinct lung and skin defects. This study expands the potential clinical heterogeneity of TARS1-related recessive disease, which should guide future clinical and genetic evaluations of patient populations.
Collapse
Affiliation(s)
| | - Allison R Cale
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | - Kira E Jonatzke
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Young N Park
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | - Guy M Lenk
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA
| | | | | | - Andrzej A Dlugosz
- Department of Dermatology, University of Michigan, Ann Arbor, MI, USA; Rogel Cancer Center, University of Michigan, Ann Arbor, MI, USA; Department of Cell and Developmental Biology, University of Michigan, Ann Arbor, MI, USA
| | - Asim A Beg
- Neuroscience Graduate Program, University of Michigan, Ann Arbor, MI, USA
| | - Miriam H Meisler
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, USA
| | - Anthony Antonellis
- Department of Human Genetics, University of Michigan, Ann Arbor, MI, USA; Department of Neurology, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
13
|
Watanabe M, Sasaki N. Mechanisms and Future Research Perspectives on Mitochondrial Diseases Associated with Isoleucyl-tRNA Synthetase Gene Mutations. Genes (Basel) 2024; 15:894. [PMID: 39062673 PMCID: PMC11276352 DOI: 10.3390/genes15070894] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 07/03/2024] [Accepted: 07/05/2024] [Indexed: 07/28/2024] Open
Abstract
Aminoacyl-tRNA synthetases are essential enzymes for the accurate translation of genetic information. IARS1 and IARS2 are isoleucyl-tRNA synthetases functioning in the cytoplasm and mitochondria, respectively, with genetic mutations in these enzymes causing diverse clinical phenotypes in specific organs and tissues. Mutations in IARS1 and IARS2 have recently been linked to mitochondrial diseases. This review aims to explore the relationship between IARS1 and IARS2 and these diseases, providing a comprehensive overview of their association with mitochondrial diseases. Mutations in IARS1 cause weak calf syndrome in cattle and mitochondrial diseases in humans, leading to growth retardation and liver dysfunction. Mutations in IARS2 are associated with Leigh syndrome, craniosynostosis and abnormal genitalia syndrome. Future research is expected to involve genetic analysis of a larger number of patients, identifying new mutations in IARS1 and IARS2, and elucidating their impact on mitochondrial function. Additionally, genetically modified mice and the corresponding phenotypic analysis will serve as powerful tools for understanding the functions of these gene products and unraveling disease mechanisms. This will likely promote the development of new therapies and preventive measures.
Collapse
Affiliation(s)
| | - Nobuya Sasaki
- Laboratory of Laboratory Animal Science and Medicine, Kitasato University, 35-1, Higashi-23, Towada 034-8628, Aomori, Japan
| |
Collapse
|
14
|
Hegarty R, Thompson RJ. Genetic aetiologies of acute liver failure. J Inherit Metab Dis 2024; 47:582-597. [PMID: 38499319 DOI: 10.1002/jimd.12733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 03/11/2024] [Accepted: 03/12/2024] [Indexed: 03/20/2024]
Abstract
Acute liver failure (ALF) is a rare, rapidly evolving, clinical syndrome with devastating consequences where definitive treatment is by emergency liver transplantation. Establishing a diagnosis can be challenging and, historically, the cause of ALF was unidentified in up to half of children. However, recent technological and clinical advances in genomic medicine have led to an increasing proportion being diagnosed with monogenic aetiologies of ALF. The conditions encountered include a diverse group of inherited metabolic disorders each with prognostic and treatment implications. Often these disorders are clinically indistinguishable and may even mimic disorders of immune regulation or red cell disorders. Rapid genomic sequencing for children with ALF is, therefore, a key component in the diagnostic work up today. This review focuses on the monogenic aetiologies of ALF.
Collapse
Affiliation(s)
- Robert Hegarty
- Paediatric Liver, GI and Nutrition Centre, King's College Hospital, London, UK
- Institute of Liver Studies, King's College London, London, UK
| | - Richard J Thompson
- Paediatric Liver, GI and Nutrition Centre, King's College Hospital, London, UK
- Institute of Liver Studies, King's College London, London, UK
| |
Collapse
|
15
|
Li SY, Feng JY, Li ZD, Liu T. Early onset and liver failure indicating poor prognosis of infant liver failure syndrome type 1. Orphanet J Rare Dis 2024; 19:225. [PMID: 38844943 PMCID: PMC11155007 DOI: 10.1186/s13023-024-03229-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 05/27/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Infantile liver failure syndrome type 1 (ILFS1, OMIM #615,438), caused by leucyl-tRNA synthase 1 (LARS1, OMIM *151,350) deficiency, is a rare autosomal-recessive disorder. The clinical manifestations, molecular-genetic features, and prognosis of LARS1 disease remain largely elusive. METHODS Three new instances of ILFS1 with confirmed variants in LARS1, encoding LARS1, were identified. Disease characteristics were summarized together with those of 33 reported cases. Kaplan-Meier analysis was performed to assess prognostic factors in ILFS1 patients. RESULTS The 3 new ILFS1 patients harbored 6 novel variants in LARS1. Among the 36 known patients, 12 died or underwent liver transplantation. The main clinical features of ILFS1 were intrauterine growth restriction (31/32 patients in whom this finding was specifically described), failure to thrive (30/31), hypoalbuminemia (32/32), microcytic anemia (32/33), acute liver failure (24/34), neurodevelopmental delay (25/30), seizures (22/29), and muscular hypotonia (13/27). No significant correlations were observed between genotype and either presence of liver failure or clinical severity of disease. Kaplan-Meier analysis indicated that age of onset < 3mo (p = 0.0015, hazard ratio = 12.29, 95% confidence interval [CI] = 3.74-40.3), like liver failure (p = 0.0343, hazard ratio = 6.57, 95% CI = 1.96-22.0), conferred poor prognosis. CONCLUSIONS Early age of presentation, like liver failure, confers poor prognosis in ILFS1. Genotype-phenotype correlations remain to be established.
Collapse
Affiliation(s)
- Shu-Yuan Li
- Department of Hepatology, The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Jia-Yan Feng
- The Department of Pathology, Children's Hospital of Fudan University, Shanghai, 201102, China
| | - Zhong-Die Li
- Department of Hepatology, The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China
| | - Teng Liu
- Department of Hepatology, The Center for Pediatric Liver Diseases, Children's Hospital of Fudan University, 399 Wanyuan Road, Shanghai, 201102, China.
| |
Collapse
|
16
|
Warasnhe K, Özçay F, Kılıç E, Sezer T, Haberal M. SCYL1 deficiency: A rare entity with challenging neurological manifestations after liver transplantation. Pediatr Transplant 2024; 28:e14661. [PMID: 38149330 DOI: 10.1111/petr.14661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 10/07/2023] [Accepted: 10/26/2023] [Indexed: 12/28/2023]
Abstract
BACKGROUND Pediatric acute liver failure (PALF) with undetermined etiology is associated with higher liver transplantation and lower spontaneous recovery (transplant-free) rates. The diagnostic odyssey in PALF cases hinders appropriate management and follow-up after liver transplantation. Advances in whole exome sequencing analysis have already been successful at identifying new genetic causes of PALF. CASE PRESENTATION We report a 17-year-old girl who underwent liver transplantation at the age of 7 months due to acute liver failure and presented later with abnormal neurological manifestations, that is, gait disturbances, dysarthria, and mental retardation that led us to the diagnosis of SCYL1 deficiency. CONCLUSION PALF cases should be screened for possible underlying genetic disorders. Genetic studies and reanalysis of whole-genome sequencing data may help identify new cases and clarify the genotype-phenotype correlation. SCYL1 deficiency should be suspected in PALF patients who develop neurological involvement after LT. Early diagnosis is vital for proper management of ALF crises in SCYL1 deficiency patients. Despite the reported favorable outcomes of ALF crises in SCYL1 deficiency, liver transplantation decision should be discussed on a case-by-case basis.
Collapse
Affiliation(s)
- Khaled Warasnhe
- Department of Pediatrics, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Figen Özçay
- Department of Pediatric Gastroenterology and Hepatology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Esra Kılıç
- Department of Pediatric Genetics, Ankara Bilkent City Hospital, Ankara, Turkey
| | - Taner Sezer
- Department of Pediatric Neurology, Başkent University Faculty of Medicine, Ankara, Turkey
| | - Mehmet Haberal
- Department of General Surgery, Başkent University Faculty of Medicine, Ankara, Turkey
| |
Collapse
|
17
|
Wang S, Liu S, Zhu Y, Zhang B, Yang Y, Li L, Sun Y, Zhang L, Fan L, Hu X, Huang C. A novel and independent survival prognostic model for OSCC: the functions and prognostic values of RNA-binding proteins. Eur Arch Otorhinolaryngol 2024; 281:397-409. [PMID: 37656222 DOI: 10.1007/s00405-023-08200-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 08/17/2023] [Indexed: 09/02/2023]
Abstract
BACKGROUND Oral squamous cell carcinoma (OSCC), exhibiting high morbidity and malignancy, is the most common type of oral cancer. The abnormal expression of RNA-binding proteins (RBPs) plays important roles in the occurrence and progression of cancer. The objective of the present study was to establish a prognostic assessment model of RBPs and to evaluate the prognosis of OSCC patients. METHODS Gene expression data in The Cancer Genome Atlas (TCGA) were analyzed by univariate Cox regression analysis model that established a novel nine RBPs, which were used to build a prognostic risk model. A multivariate Cox proportional regression model and the survival analysis were used to evaluate the prognostic risk model. Moreover, the receive operator curve (ROC) analysis was tested further the efficiency of prognostic risk model based on data from TCGA database and Gene Expression Omnibus (GEO). RESULTS Nine RBPs' signatures (ACO1, G3BP1, NMD3, RNGTT, ZNF385A, SARS, CARS2, YARS and SMAD6) with prognostic value were identified in OSCC patients. Subsequently, the patients were further categorized into high-risk group and low-risk in the overall survival (OS) and disease-free survival (DFS), and external validation dataset. ROC analysis was significant for both the TCGA and GEO. Moreover, GSEA revealed that patients in the high-risk group significantly enriched in many critical pathways correlated with tumorigenesis than the low, including cell cycle, adheres junctions, oocyte meiosis, spliceosome, ERBB signaling pathway and ubiquitin-mediated proteolysis. CONCLUSIONS Collectively, we developed and validated a novel robust nine RBPs for OSCC prognosis prediction. The nine RBPs could serve as an independent and reliable prognostic biomarker and guiding clinical therapy for OSCC patients.
Collapse
Affiliation(s)
- Shanshan Wang
- Shenzhen Stomatology Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Shuang Liu
- Shenzhen Luohu People's Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Yaomin Zhu
- Shenzhen Stomatology Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Baorong Zhang
- Department of Stomatology, University of Chinese Academy of Sciences Shenzhen Hospital, Songbai Road 4253, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Yongtao Yang
- Shenzhen Stomatology Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Limei Li
- Shenzhen Stomatology Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Yingying Sun
- Shenzhen Stomatology Hospital, Shenzhen, 518001, Guangdong, People's Republic of China
| | - Long Zhang
- Department of Stomatology, University of Chinese Academy of Sciences Shenzhen Hospital, Songbai Road 4253, Shenzhen, 518107, Guangdong, People's Republic of China
| | - Lina Fan
- Department of Stomatology, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, 350025, Fujian, China
| | - Xuegang Hu
- Department of Stomatology, University of Chinese Academy of Sciences Shenzhen Hospital, Songbai Road 4253, Shenzhen, 518107, Guangdong, People's Republic of China.
| | - Chunyu Huang
- Department of Stomatology, University of Chinese Academy of Sciences Shenzhen Hospital, Songbai Road 4253, Shenzhen, 518107, Guangdong, People's Republic of China.
- Medical Affairs Department, University of Chinese Academy of Sciences-Shenzhen Hospital, Songbai Road 4253, Shenzhen, 518107, Guangdong, China.
| |
Collapse
|
18
|
Jiang J, Feng Y, Tang Q, Zhao C, Guo M, Wu J, Guo R, Lu H, Sun X, Gao J, Xue H. Novel IARS1 variants cause syndromic developmental disorder with epilepsy in a Chinese patient and the literature review. Mol Genet Genomic Med 2024; 12:e2326. [PMID: 38014478 PMCID: PMC10767687 DOI: 10.1002/mgg3.2326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 10/20/2023] [Accepted: 11/14/2023] [Indexed: 11/29/2023] Open
Abstract
BACKGROUND Isoleucinyl-tRNA synthetase (IARS) is encoded by the IARS1 gene and catalyzes the binding of isoleucine to specific tRNA. OBJECTIVE This study aims to investigate the pathogenicity of novel IARS1 variants and the genotype-phenotype association, in order to expand the spectrum of pathogenic variants and phenotypes of IARS1-related disease and provide new evidence for the phenotypic spectrum of IARS1 variants. METHODS Clinical data of the proband were collected, and trio whole-exome sequencing (WES) was performed on the proband and the parents. Candidate variants were validated using Sanger sequencing. Bioinformatics software was utilized to analyze the functional consequences of identified variants and predict their potential deleteriousness. RESULTS A 17-month-old female patient presented with microcephaly, left external ear malformation, decreased muscle strength and tone in all limbs, epileptic seizures, global developmental delay, and developmental regression. Trio WES identified compound heterozygous variants in the IARS1 gene, c.120-1G>A and c.2164C>A, which were novel pathogenic and likely pathogenic variants, respectively. The phenotype of developmental regression has not been reported before. Only one patient with IARS1 compound heterozygous variants has been reported in the world to have an epileptic phenotype, and this is the second patient with an epileptic phenotype. Bioinformatics analysis revealed that the splicing variant disrupted the canonical splice donor site, while the missense variant altered the local electrostatics of the IARS1 protein surface, potentially leading to functional abnormalities. CONCLUSION This study identified novel IARS1 variants and the phenotype of developmental regression, expanding the spectrum of pathogenic variants and phenotypes of IARS1-related diseases and providing new evidence for the rare phenotype of epileptic seizures caused by IARS1 variants.
Collapse
Affiliation(s)
- Jinsong Jiang
- Department of Paediatric MedicineShanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
- Department of Cytogenetic Laboratory, Children's Hospital of ShanxiWomen Health Center of Shanxi, Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Yu Feng
- Department of Paediatric MedicineShanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Qiaoyin Tang
- Department of Paediatric MedicineShanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Chenyue Zhao
- Department of Paediatric MedicineShanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Min Guo
- Department of Paediatric MedicineShanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Jianrui Wu
- Department of Cytogenetic Laboratory, Children's Hospital of ShanxiWomen Health Center of Shanxi, Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Rong Guo
- Department of Cytogenetic Laboratory, Children's Hospital of ShanxiWomen Health Center of Shanxi, Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Hongyong Lu
- Department of Cytogenetic Laboratory, Children's Hospital of ShanxiWomen Health Center of Shanxi, Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Xiayu Sun
- Department of Cytogenetic Laboratory, Children's Hospital of ShanxiWomen Health Center of Shanxi, Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Jingbo Gao
- Department of Cytogenetic Laboratory, Children's Hospital of ShanxiWomen Health Center of Shanxi, Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| | - Huiqin Xue
- Department of Cytogenetic Laboratory, Children's Hospital of ShanxiWomen Health Center of Shanxi, Affiliated Hospital of Shanxi Medical UniversityTaiyuanShanxiPeople's Republic of China
| |
Collapse
|
19
|
Dunlea E, Crushell E, Cotter M, Blau N, Ferreira CR. Clinical and biochemical footprints of inherited metabolic disease. XVI. Hematological abnormalities. Mol Genet Metab 2023; 140:107735. [PMID: 37989003 PMCID: PMC11752444 DOI: 10.1016/j.ymgme.2023.107735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/08/2023] [Accepted: 11/09/2023] [Indexed: 11/23/2023]
Abstract
Many classical inherited metabolic diseases (IMDs) are associated with significant hematological complications such as anemia or thrombosis. While these may not be the prominent presenting feature of these conditions, management of these issues is important for optimal outcomes in people with IMDs. Some disorders that are included in the nosology of inherited metabolic disorders, such as inherited disorders of red cell energy metabolism, have purely hematological features, and have typically been cared for by a hematologist. In the 16th issue of the Footprints series, we identified 265 IMDs associated with hematological abnormalities. We review the major hematological manifestations of IMDs, suggest further investigation of hematological findings, and discuss treatment options available for specific hematological complications of IMDs.
Collapse
Affiliation(s)
- Eoghan Dunlea
- Dept of Haematology, Children's Health Ireland, Temple Street, Dublin, Ireland; School of Medicine, Trinity College, Dublin, Ireland.
| | - Ellen Crushell
- National Centre for Inherited Metabolic Disorders, Children's Health Ireland, Temple Street, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland
| | - Melanie Cotter
- Dept of Haematology, Children's Health Ireland, Temple Street, Dublin, Ireland; School of Medicine, University College Dublin, Dublin, Ireland.
| | - Nenad Blau
- Division of Metabolism, University Children's Hospital, Zürich, Switzerland.
| | - Carlos R Ferreira
- National Human Genome Research Institute, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
20
|
彭 文, 朱 燕, 王 来, 陆 炜, 杨 琳, 朱 丽. [A case of interstitial lung and liver disease caused by MARS1 gene mutation]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2023; 25:1186-1190. [PMID: 37990466 PMCID: PMC10672959 DOI: 10.7499/j.issn.1008-8830.2307050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 09/13/2023] [Indexed: 11/23/2023]
Abstract
The patient is a female infant, 4 months and 9 days old, who was admitted to the hospital due to recurrent fever, cough, and hepatomegaly for over a month. The patient was a healthy full-term infant with a normal birth history. At 2 months and 22 days after birth, she developed recurrent fever, cough, and respiratory distress. Chest imaging revealed diffuse bilateral lung lesions, and fiberoptic bronchoscopy showed interstitial changes in both lungs. These suggested the presence of interstitial lung disease. The patient also presented with hepatomegaly, anemia, hyperlipidemia, hypothyroidism, and malnutrition. Genetic testing indicated compound heterozygous variations in the MARS1 gene. This mutation can cause interstitial lung and liver disease, which is a severe rare disorder that typically manifests in infancy or early childhood. It is inherited in an autosomal recessive manner and characterized by early-onset respiratory insufficiency and liver disease in infants or young children. Since its first reported case in 2013, as of June 2023, only 38 related cases have been reported worldwide. This article reports the multidisciplinary diagnosis and treatment of interstitial lung and liver disease in an infant caused by MARS1 gene mutation.
Collapse
Affiliation(s)
| | | | | | - 炜 陆
- 国家儿童医学中心/复旦大学附属儿科医院内分泌遗传代谢科上海201102
| | - 琳 杨
- 国家儿童医学中心/复旦大学附属儿科医院内分泌遗传代谢科上海201102
| | | |
Collapse
|
21
|
Saettini F, Guerra F, Fazio G, Bugarin C, McMillan HJ, Ohtake A, Ardissone A, Itoh M, Giglio S, Cappuccio G, Giardino G, Romano R, Quadri M, Gasperini S, Moratto D, Chiarini M, Akira I, Fukuhara Y, Hayakawa I, Okazaki Y, Mauri M, Piazza R, Cazzaniga G, Biondi A. Antibody Deficiency in Patients with Biallelic KARS1 Mutations. J Clin Immunol 2023; 43:2115-2125. [PMID: 37770806 DOI: 10.1007/s10875-023-01584-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Accepted: 09/12/2023] [Indexed: 09/30/2023]
Abstract
Biallelic KARS1 mutations cause KARS-related diseases, a rare syndromic condition encompassing central and peripheral nervous system impairment, heart and liver disease, and deafness. KARS1 encodes the t-RNA synthase of lysine, an aminoacyl-tRNA synthetase, involved in different physiological mechanisms (such as angiogenesis, post-translational modifications, translation initiation, autophagy and mitochondrial function). Although patients with immune-hematological abnormalities have been individually described, results have not been collectively discussed and functional studies investigating how KARS1 mutations affect B cells have not been performed. Here, we describe one patient with severe developmental delay, sensoneurinal deafness, acute disseminated encephalomyelitis, hypogammaglobulinemia and recurrent infections. Pathogenic biallelic KARS1 variants (Phe291Val/ Pro499Leu) were associated with impaired B cell metabolism (decreased mitochondrial numbers and activity). All published cases of KARS-related diseases were identified. The corresponding authors and researchers involved in the diagnosis of inborn errors of immunity or genetic syndromes were contacted to obtain up-to-date clinical and immunological information. Seventeen patients with KARS-related diseases were identified. Recurrent/severe infections (9/17) and B cell abnormalities (either B cell lymphopenia [3/9], hypogammaglobulinemia [either IgG, IgA or IgM; 6/15] or impaired vaccine responses [4/7]) were frequently reported. Immunoglobulin replacement therapy was given in five patients. Full immunological assessment is warranted in these patients, who may require detailed investigation and specific supportive treatment.
Collapse
Affiliation(s)
- Francesco Saettini
- Centro Tettamanti, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy.
| | - Fabiola Guerra
- Pediatria, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- Dipartimento Di Medicina E Chirurgia, Università Degli Studi Milano-Bicocca, Milan, Italy
| | - Grazia Fazio
- Centro Tettamanti, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Cristina Bugarin
- Centro Tettamanti, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Hugh J McMillan
- Children's Hospital of Eastern Ontario Research Institute, University of Ottawa, Ottawa, ON, Canada
| | - Akira Ohtake
- Department of Clinical Genomics & Pediatrics, Saitama Medical University, Moroyama, Saitama, Japan
| | - Anna Ardissone
- Child Neurology, "Fondazione IRCCS IstitutoNeurologico Carlo Besta, Via Celoria 11, 20133, Milan, Italy
| | - Masayuki Itoh
- Department of Mental Retardation and Birth Defect Research, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Sabrina Giglio
- Unit of Medical Genetics, Department of Medical Sciences and Public Health, University of Cagliari, Cagliari, Italy
| | - Gerarda Cappuccio
- Department of Translational Medical Sciences, Section of Pediatrics, Federico II University of Naples, Naples, Italy
- Current address: Baylor College of Medicine, Houston, TX, USA
| | - Giuliana Giardino
- Department of Translational Medical Sciences, Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Roberta Romano
- Department of Translational Medical Sciences, Section of Pediatrics, Federico II University of Naples, Naples, Italy
| | - Manuel Quadri
- Centro Tettamanti, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Serena Gasperini
- Pediatria, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
| | - Daniele Moratto
- Flow Cytometry Unit, Clinical ChemistryLaboratory, ASST Spedali Civili, Brescia, Italy
| | - Marco Chiarini
- Flow Cytometry Unit, Clinical ChemistryLaboratory, ASST Spedali Civili, Brescia, Italy
| | - Ishiguro Akira
- Center for Postgraduate Education and Training, National Center for Child Health and Development (NCCHD), Tokyo, Japan
- Division of Hematology, National Center for Child Health and Development (NCCHD), Tokyo, Japan
| | - Yasuyuki Fukuhara
- Division of Medical Genetics, National Center for Child Health and Development, Tokyo, Japan
| | - Itaru Hayakawa
- Division of Neurology, National Center for Child Health and Development (NCCHD), Tokyo, Japan
| | - Yasushi Okazaki
- Division of Neurology, National Center for Child Health and Development (NCCHD), Tokyo, Japan
- Diagnostics and Therapeutic of Intractable Diseases, Intractable Disease Research Center, Graduate School of Medicine, Juntendo University, Tokyo, Japan
| | - Mario Mauri
- Dipartimento Di Medicina E Chirurgia, Università Degli Studi Milano-Bicocca, Milan, Italy
| | - Rocco Piazza
- Ematologia, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Gianni Cazzaniga
- Centro Tettamanti, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- Dipartimento Di Medicina E Chirurgia, Università Degli Studi Milano-Bicocca, Milan, Italy
| | - Andrea Biondi
- Centro Tettamanti, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- Pediatria, Fondazione IRCCS San Gerardo Dei Tintori, Monza, Italy
- Dipartimento Di Medicina E Chirurgia, Università Degli Studi Milano-Bicocca, Milan, Italy
| |
Collapse
|
22
|
Tijaro-Bulla S, Nyandwi SP, Cui H. Physiological and engineered tRNA aminoacylation. WILEY INTERDISCIPLINARY REVIEWS. RNA 2023; 14:e1789. [PMID: 37042417 DOI: 10.1002/wrna.1789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/23/2022] [Revised: 03/11/2023] [Accepted: 03/21/2023] [Indexed: 04/13/2023]
Abstract
Aminoacyl-tRNA synthetases form the protein family that controls the interpretation of the genetic code, with tRNA aminoacylation being the key chemical step during which an amino acid is assigned to a corresponding sequence of nucleic acids. In consequence, aminoacyl-tRNA synthetases have been studied in their physiological context, in disease states, and as tools for synthetic biology to enable the expansion of the genetic code. Here, we review the fundamentals of aminoacyl-tRNA synthetase biology and classification, with a focus on mammalian cytoplasmic enzymes. We compile evidence that the localization of aminoacyl-tRNA synthetases can be critical in health and disease. In addition, we discuss evidence from synthetic biology which made use of the importance of subcellular localization for efficient manipulation of the protein synthesis machinery. This article is categorized under: RNA Processing Translation > Translation Regulation RNA Processing > tRNA Processing RNA Export and Localization > RNA Localization.
Collapse
Affiliation(s)
| | | | - Haissi Cui
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
23
|
Kazakova E, Téllez-Martínez JA, Flores-Lagunes L, Sosa-Ortiz AL, Carillo-Sánchez K, Molina-Garay C, González-Domínguez CA, Jimenez-Olivares M, Fernandez-Valverde F, Vargas-Cañas ES, Vázquez-Memije ME, Garcia-Latorre EA, Martínez-Duncker I, Alaez-Verson C. Uterus infantilis: a novel phenotype associated with AARS2 new genetic variants. A case report. Front Neurol 2023; 14:878446. [PMID: 37456626 PMCID: PMC10343430 DOI: 10.3389/fneur.2023.878446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Accepted: 05/10/2023] [Indexed: 07/18/2023] Open
Abstract
Objectives To report the first Mexican case with two novel AARS2 mutations causing primary ovarian failure, uterus infantilis, and early-onset dementia secondary to leukoencephalopathy. Methods Detailed clinical, clinimetric, neuroimaging features, muscle biopsy with biochemical assays of the main oxidative phosphorylation complexes activities, and molecular studies were performed on samples from a Mexican female. Results We present a 41-year-old female patient with learning difficulties since childhood and primary amenorrhea who developed severe cognitive, motor, and behavioral impairment in early adulthood. Neuroimaging studies revealed frontal leukoencephalopathy with hypometabolism at the fronto-cerebellar cortex and caudate nucleus. Uterus infantilis was detected on ultrasound study. Clinical exome sequencing identified two novel variants, NM_020745:c.2864G>A (p.W955*) and NM_020745:c.1036C>A (p.P346T, p.P346Wfs*18), in AARS2. Histopathological and biochemical studies on muscle biopsy revealed mitochondrial disorder with cytochrome C oxidase (COX) deficiency. Conclusions Several adult-onset cases of leukoencephalopathy and ovarian failure associated with AARS2 variants have been reported. To our best knowledge, none of them showed uterus infantilis. Here we enlarge the genetic and phenotypic spectrum of AARS2-related dementia with leukoencephalopathy and ovarian failure and contribute with detailed clinical, clinometric, neuroimaging, and molecular studies to disease and novel molecular variants characterization.
Collapse
Affiliation(s)
- Ekaterina Kazakova
- Centro de Diagnóstico en Metabolismo Energético y Medicina Mitocondrial, Mexico City, Mexico
| | - José Alberto Téllez-Martínez
- Clínica de Cognición, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Leonardo Flores-Lagunes
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Ana Luisa Sosa-Ortiz
- Clínica de Cognición, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Karol Carillo-Sánchez
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Carolina Molina-Garay
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Carlos Alberto González-Domínguez
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Morelos, Mexico
| | - Marco Jimenez-Olivares
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Francisca Fernandez-Valverde
- Laboratorio de Patología Experimental, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | - Edwin Steven Vargas-Cañas
- Clínica de Nervio y Músculo, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, Mexico City, Mexico
| | | | | | - Iván Martínez-Duncker
- Laboratorio de Glicobiología Humana y Diagnóstico Molecular, Centro de Investigación en Dinámica Celular, Instituto de Investigación en Ciencias Básicas y Aplicadas, Universidad Autónoma del Estado de Morelos, Morelos, Mexico
| | - Carmen Alaez-Verson
- Laboratorio de Diagnóstico Genómico, Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| |
Collapse
|
24
|
Kalotay E, Klugmann M, Housley GD, Fröhlich D. Dominant aminoacyl-tRNA synthetase disorders: lessons learned from in vivo disease models. Front Neurosci 2023; 17:1182845. [PMID: 37274211 PMCID: PMC10234151 DOI: 10.3389/fnins.2023.1182845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/05/2023] [Indexed: 06/06/2023] Open
Abstract
Aminoacyl-tRNA synthetases (ARSs) play an essential role in protein synthesis, being responsible for ligating tRNA molecules to their corresponding amino acids in a reaction known as 'tRNA aminoacylation'. Separate ARSs carry out the aminoacylation reaction in the cytosol and in mitochondria, and mutations in almost all ARS genes cause pathophysiology most evident in the nervous system. Dominant mutations in multiple cytosolic ARSs have been linked to forms of peripheral neuropathy including Charcot-Marie-Tooth disease, distal hereditary motor neuropathy, and spinal muscular atrophy. This review provides an overview of approaches that have been employed to model each of these diseases in vivo, followed by a discussion of the existing animal models of dominant ARS disorders and key mechanistic insights that they have provided. In summary, ARS disease models have demonstrated that loss of canonical ARS function alone cannot fully account for the observed disease phenotypes, and that pathogenic ARS variants cause developmental defects within the peripheral nervous system, despite a typically later onset of disease in humans. In addition, aberrant interactions between mutant ARSs and other proteins have been shown to contribute to the disease phenotypes. These findings provide a strong foundation for future research into this group of diseases, providing methodological guidance for studies on ARS disorders that currently lack in vivo models, as well as identifying candidate therapeutic targets.
Collapse
Affiliation(s)
- Elizabeth Kalotay
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gary D. Housley
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Dominik Fröhlich
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
25
|
Kalotay E, Klugmann M, Housley GD, Fröhlich D. Recessive aminoacyl-tRNA synthetase disorders: lessons learned from in vivo disease models. Front Neurosci 2023; 17:1182874. [PMID: 37274208 PMCID: PMC10234152 DOI: 10.3389/fnins.2023.1182874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2023] [Accepted: 04/17/2023] [Indexed: 06/06/2023] Open
Abstract
Protein synthesis is a fundamental process that underpins almost every aspect of cellular functioning. Intriguingly, despite their common function, recessive mutations in aminoacyl-tRNA synthetases (ARSs), the family of enzymes that pair tRNA molecules with amino acids prior to translation on the ribosome, cause a diverse range of multi-system disorders that affect specific groups of tissues. Neurological development is impaired in most ARS-associated disorders. In addition to central nervous system defects, diseases caused by recessive mutations in cytosolic ARSs commonly affect the liver and lungs. Patients with biallelic mutations in mitochondrial ARSs often present with encephalopathies, with variable involvement of peripheral systems. Many of these disorders cause severe disability, and as understanding of their pathogenesis is currently limited, there are no effective treatments available. To address this, accurate in vivo models for most of the recessive ARS diseases are urgently needed. Here, we discuss approaches that have been taken to model recessive ARS diseases in vivo, highlighting some of the challenges that have arisen in this process, as well as key results obtained from these models. Further development and refinement of animal models is essential to facilitate a better understanding of the pathophysiology underlying recessive ARS diseases, and ultimately to enable development and testing of effective therapies.
Collapse
Affiliation(s)
- Elizabeth Kalotay
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Matthias Klugmann
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
- Research Beyond Borders, Boehringer Ingelheim Pharma GmbH & Co. KG, Biberach an der Riss, Germany
| | - Gary D. Housley
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| | - Dominik Fröhlich
- Translational Neuroscience Facility and Department of Physiology, School of Biomedical Sciences, University of New South Wales, Sydney, NSW, Australia
| |
Collapse
|
26
|
Mann JP, Lenz D, Stamataki Z, Kelly D. Common mechanisms in pediatric acute liver failure. Trends Mol Med 2023; 29:228-240. [PMID: 36496278 DOI: 10.1016/j.molmed.2022.11.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2022] [Revised: 11/06/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Acute liver failure (ALF) is a rare but potentially fatal disease in children. The etiology is multifactorial, including infection, autoimmune, and genetic disorders, as well as indeterminate hepatitis, which has a higher requirement for liver transplantation. Activation of the innate and adaptive immune systems leads to hepatocyte-specific injury which is mitigated by T regulatory cell activation. Recovery of the native liver depends on activation of apoptotic and regenerative pathways, including the integrated stress response (ISR; e.g., PERK), p53, and HNF4α. Loss-of-function mutations in these pathways cause recurrent ALF in response to non-hepatotropic viruses. Deeper understanding of these mechanisms will lead to improved diagnosis, management, and outcomes for pediatric ALF.
Collapse
Affiliation(s)
- Jake P Mann
- Liver Unit, Birmingham Women's and Children's Hospital, and University of Birmingham, Birmingham, UK
| | - Dominic Lenz
- Division of Neuropediatrics and Pediatric Metabolic Medicine, Center for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Zania Stamataki
- Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK
| | - Deirdre Kelly
- Liver Unit, Birmingham Women's and Children's Hospital, and University of Birmingham, Birmingham, UK; Centre for Liver and Gastrointestinal Research, Institute of Immunology and Immunotherapy, University of Birmingham, Birmingham, UK.
| |
Collapse
|
27
|
Nathan N, Griese M, Michel K, Carlens J, Gilbert C, Emiralioglu N, Torrent-Vernetta A, Marczak H, Willemse B, Delestrain C, Epaud R. Diagnostic workup of childhood interstitial lung disease. Eur Respir Rev 2023; 32:32/167/220188. [PMID: 36813289 PMCID: PMC9945877 DOI: 10.1183/16000617.0188-2022] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2022] [Accepted: 12/18/2022] [Indexed: 02/24/2023] Open
Abstract
Childhood interstitial lung diseases (chILDs) are rare and heterogeneous diseases with significant morbidity and mortality. An accurate and quick aetiological diagnosis may contribute to better management and personalised treatment. On behalf of the European Respiratory Society Clinical Research Collaboration for chILD (ERS CRC chILD-EU), this review summarises the roles of the general paediatrician, paediatric pulmonologists and expert centres in the complex diagnostic workup. Each patient's aetiological chILD diagnosis must be reached without prolonged delays in a stepwise approach from medical history, signs, symptoms, clinical tests and imaging, to advanced genetic analysis and specialised procedures including bronchoalveolar lavage and biopsy, if necessary. Finally, as medical progress is fast, the need to revisit a diagnosis of "undefined chILD" is stressed.
Collapse
Affiliation(s)
- Nadia Nathan
- AP-HP, Sorbonne Université, Pediatric Pulmonology Department and Reference Center for Rare Lung Disease RespiRare, Armand Trousseau Hospital, Paris, France .,Sorbonne Université, Inserm UMR_S933 Laboratory of Childhood Genetic Diseases, Armand Trousseau Hospital, Paris, France
| | - Matthias Griese
- Department of Paediatric Pneumology, Dr von Hauner Children's Hospital, German Centre for Lung Research, University of Munich, Munich, Germany
| | - Katarzyna Michel
- Department of Paediatric Pneumology, Dr von Hauner Children's Hospital, German Centre for Lung Research, University of Munich, Munich, Germany
| | - Julia Carlens
- Clinic for Pediatric Pneumology, Hannover Medical School, Hannover, Germany
| | - Carlee Gilbert
- Institute of Population Health, University of Liverpool, Liverpool, UK
| | - Nagehan Emiralioglu
- Department of Pediatric Pulmonology, Hacettepe University Faculty of Medicine, Ankara, Turkey
| | - Alba Torrent-Vernetta
- Pediatric Allergy and Pulmonology Section, Department of Pediatrics, Vall d’Hebron Hospital Universitari, Vall d’Hebron Barcelona Hospital Campus, Universitat Autònoma de Barcelona, Barcelona, Spain,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Honorata Marczak
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
| | - Brigitte Willemse
- Department of Pediatric Pneumology and Allergy, Medical University of Warsaw, Warsaw, Poland
| | - Céline Delestrain
- Department of Pediatric Pulmonology and Pediatric Allergology, Beatrix Children's Hospital, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands,Centre Hospitalier Intercommunal de Créteil, Service de Pédiatrie Générale, Créteil, France,Centre des Maladies Respiratoires Rares (RESPIRARE®), CRCM, Créteil, France
| | - Ralph Epaud
- Centre Hospitalier Intercommunal de Créteil, Service de Pédiatrie Générale, Créteil, France,Centre des Maladies Respiratoires Rares (RESPIRARE®), CRCM, Créteil, France,University Paris Est Créteil, INSERM, IMRB, Créteil, France
| |
Collapse
|
28
|
Li Y, Liu G, Yu F, Jiang Y. Successful treatment of rapid progressive interstitial lung disease in a case of anti-Zo antibody positive anti-synthetase syndrome. Int J Rheum Dis 2023; 26:370-375. [PMID: 36269556 DOI: 10.1111/1756-185x.14471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 09/26/2022] [Accepted: 10/03/2022] [Indexed: 02/01/2023]
Abstract
BACKGROUND Anti-synthetase syndrome (ASS) is a chronic multisystemic autoimmune disease characterized by detectable anti-aminoacyl-transfer-RNA antibodies. Interstitial lung disease (ILD) in anti-synthetase syndrome patients is often severe and rapidly progressive. Anti-Zo (phenylalanyl) antibody is reported rarely in ASS. Therefore, the appropriate treatment of anti-Zo positive ASS is unclear. CASE PRESENTATION Here we present a case of anti-Zo-positive ASS with rapid progressive ILD (RP-ILD) in a Chinese patient successfully treated with a combination of systemic corticosteroids and tacrolimus. CONCLUSION We reviewed 13 anti-Zo-positive ASS patients (including our case) and summarized clinical features that have some differences with other ASS. Anti-Zo-positive ASS is a rare autoimmune disease with a high burden of ILD, is often severe and rapidly progressive. Corticosteroids with tacrolimus may improve patient outcomes in anti-Zo antibody positive ASS with RP-ILD.
Collapse
Affiliation(s)
- Yongxia Li
- Department of Respiratory and Critical Care Medicine, The University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Gang Liu
- Department of Critical Care Medicine, The University-Town Hospital of Chongqing Medical University, Chongqing, People's Republic of China
| | - Fengjiao Yu
- Department of Respiratory and Critical Care Medicine, The University-Town Hospital of Chongqing Medical University, Chongqing, China
| | - Yu Jiang
- Department of Respiratory and Critical Care Medicine, The University-Town Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
29
|
Ait-El-Mkadem Saadi S, Kaphan E, Morales Jaurrieta A, Fragaki K, Chaussenot A, Bannwarth S, Maues De Paula A, Paquis-Flucklinger V, Rouzier C. Splicing variants in NARS2 are associated with milder phenotypes and intra-familial variability. Eur J Med Genet 2022; 65:104643. [DOI: 10.1016/j.ejmg.2022.104643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 10/07/2022] [Accepted: 10/10/2022] [Indexed: 11/03/2022]
|
30
|
Verdura E, Senger B, Raspall-Chaure M, Schlüter A, Launay N, Ruiz M, Casasnovas C, Rodriguez-Palmero A, Macaya A, Becker HD, Pujol A. Loss of seryl-tRNA synthetase ( SARS1) causes complex spastic paraplegia and cellular senescence. J Med Genet 2022; 59:1227-1233. [PMID: 36041817 PMCID: PMC9691831 DOI: 10.1136/jmg-2022-108529] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 07/25/2022] [Indexed: 01/12/2023]
Abstract
BACKGROUND Aminoacyl-tRNA synthetases (ARS) are key enzymes catalysing the first reactions in protein synthesis, with increasingly recognised pleiotropic roles in tumourgenesis, angiogenesis, immune response and lifespan. Germline mutations in several ARS genes have been associated with both recessive and dominant neurological diseases. Recently, patients affected with microcephaly, intellectual disability and ataxia harbouring biallelic variants in the seryl-tRNA synthetase encoded by seryl-tRNA synthetase 1 (SARS1) were reported. METHODS We used exome sequencing to identify the causal variant in a patient affected by complex spastic paraplegia with ataxia, intellectual disability, developmental delay and seizures, but without microcephaly. Complementation and serylation assays using patient's fibroblasts and an Saccharomyces cerevisiae model were performed to examine this variant's pathogenicity. RESULTS A de novo splice site deletion in SARS1 was identified in our patient, resulting in a 5-amino acid in-frame insertion near its active site. Complementation assays in S. cerevisiae and serylation assays in both yeast strains and patient fibroblasts proved a loss-of-function, dominant negative effect. Fibroblasts showed an abnormal cell shape, arrested division and increased beta-galactosidase staining along with a senescence-associated secretory phenotype (raised interleukin-6, p21, p16 and p53 levels). CONCLUSION We refine the phenotypic spectrum and modes of inheritance of a newly described, ultrarare neurodevelopmental disorder, while unveiling the role of SARS1 as a regulator of cell growth, division and senescence.
Collapse
Affiliation(s)
- Edgard Verdura
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Catalonia, Spain,Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | | | - Miquel Raspall-Chaure
- Pediatric Neurology Research Group, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain,Department of Paediatric Neurology, Vall d’Hebron University Hospital, 08035, Barcelona, Catalonia, Spain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Catalonia, Spain,Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Nathalie Launay
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Catalonia, Spain,Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Catalonia, Spain,Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain
| | - Carlos Casasnovas
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Catalonia, Spain,Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain,Hospital Universitari de Bellvitge, Barcelona, Spain
| | - Agustí Rodriguez-Palmero
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Catalonia, Spain,Pediatrics, Hospital Germans Trias i Pujol, Barcelona, Spain
| | - Alfons Macaya
- Pediatric Neurology Research Group, Vall d’Hebron University Hospital, Universitat Autònoma de Barcelona, 08035, Barcelona, Catalonia, Spain,Department of Paediatric Neurology, Vall d’Hebron University Hospital, 08035, Barcelona, Catalonia, Spain,Institut de Neurociències, Universitat Autònoma de Barcelona, 08193, Barcelona, Catalonia, Spain
| | | | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Bellvitge Biomedical Research Institute (IDIBELL), L'Hospitalet de Llobregat, 08908, Barcelona, Catalonia, Spain,Centre for Biomedical Research on Rare Diseases (CIBERER), Instituto de Salud Carlos III, Madrid, Spain,Catalan Institution of Research and Advanced Studies (ICREA), 08010, Barcelona, Catalonia, Spain
| |
Collapse
|
31
|
Turvey AK, Horvath GA, Cavalcanti ARO. Aminoacyl-tRNA synthetases in human health and disease. Front Physiol 2022; 13:1029218. [PMID: 36330207 PMCID: PMC9623071 DOI: 10.3389/fphys.2022.1029218] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Accepted: 10/04/2022] [Indexed: 11/29/2022] Open
Abstract
The Aminoacyl-tRNA Synthetases (aaRSs) are an evolutionarily ancient family of enzymes that catalyze the esterification reaction linking a transfer RNA (tRNA) with its cognate amino acid matching the anticodon triplet of the tRNA. Proper functioning of the aaRSs to create aminoacylated (or “charged”) tRNAs is required for efficient and accurate protein synthesis. Beyond their basic canonical function in protein biosynthesis, aaRSs have a surprisingly diverse array of non-canonical functions that are actively being defined. The human genome contains 37 genes that encode unique aaRS proteins. To date, 56 human genetic diseases caused by damaging variants in aaRS genes have been described: 46 are autosomal recessive biallelic disorders and 10 are autosomal dominant monoallelic disorders. Our appreciation of human diseases caused by damaging genetic variants in the aaRSs has been greatly accelerated by the advent of next-generation sequencing, with 89% of these gene discoveries made since 2010. In addition to these genetic disorders of the aaRSs, anti-synthetase syndrome (ASSD) is a rare autoimmune inflammatory myopathy that involves the production of autoantibodies that disrupt aaRS proteins. This review provides an overview of the basic biology of aaRS proteins and describes the rapidly growing list of human diseases known to be caused by genetic variants or autoimmune targeting that affect both the canonical and non-canonical functions of these essential proteins.
Collapse
Affiliation(s)
- Alexandra K. Turvey
- Department of Biology, Pomona College, Claremont, CA, United States
- *Correspondence: Alexandra K. Turvey,
| | - Gabriella A. Horvath
- Division of Biochemical Genetics, Department of Pediatrics, University of British Columbia, BC Children’s Hospital, Vancouver, BC, Canada
- Adult Metabolic Diseases Clinic, Vancouver General Hospital, Vancouver, BC, Canada
| | | |
Collapse
|
32
|
Karimzadeh P, Rezakhani S, Miryounesi M, Alijanpour S. Neurodegenerative disorder and diffuse brain calcifications due to FARSB mutation in two siblings. Clin Case Rep 2022; 10:e6195. [PMID: 35937029 PMCID: PMC9347330 DOI: 10.1002/ccr3.6195] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 06/29/2022] [Accepted: 07/16/2022] [Indexed: 11/24/2022] Open
Abstract
Pathogenic mutations in the FARSB gene are associated with neurodevelopmental disorder involving the brain, liver, and lungs. We report genetic analysis of a family including two affected members with this disorder, which revealed a homozygous pathogenic missense variant, FARSB: NM_005687.4:c.853G > A:p.E285K in both affected patients. The parents were heterozygous for this variant.
Collapse
Affiliation(s)
- Parvaneh Karimzadeh
- Pediatric Neurology, Pediatric Neurology Research Center, Mofid Children's HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Sepideh Rezakhani
- Pediatric Neurology, Pediatric Neurology Research Center, Mofid Children's HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Mohammad Miryounesi
- Medical Genetics, Genomic Research Center, Taleghani HospitalShahid Beheshti University of Medical SciencesTehranIran
| | - Sahar Alijanpour
- Medical Genetics, Genomic Research Center, Taleghani HospitalShahid Beheshti University of Medical SciencesTehranIran
| |
Collapse
|
33
|
Zou TT, Sun HQ, Zhu Y, He TT, Ling WW, Zhu HM, Lin ZY, Liu YY, Liu SL, Wang H, Zhang XM. Compound heterozygous variations in IARS1 cause recurrent liver failure and growth retardation in a Chinese patient: a case report. BMC Pediatr 2022; 22:329. [PMID: 35668413 PMCID: PMC9172121 DOI: 10.1186/s12887-022-03371-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 05/18/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aminoacyl-tRNA synthetases (ARSs) are enzymes responsible for attaching amino acids to tRNA, which enables protein synthesis. Mutations in isoleucyl-tRNA synthetase (IARS1) have recently been reported to be a genetic cause for growth retardation, intellectual disability, muscular hypotonia, and infantile hepatopathy (GRIDHH). CASE PRESENTATION In this study, we reported an additional case of compound heterozygous missense variations c.701 T > C (p.L234P) and c.1555C > T (p.R519C) in IARS1, which were identified using medical exome sequencing; c.701 T > C (p.L234P) was a novel variant, and c.1555C > T (p.R519C) was found in GnomAD. Unlike other reported patients, this individual presented prominently with recurrent liver failure, which led to her death at an early age of 19 months. She also had significant growth retardation, muscular hypotonia, chubby and flabby face, recurrent loose stools, and abnormal brain computed tomography (CT), while zinc deficiency and hearing loss were not present. Studies in zebrafish embryo modeling recapitulated some of the key phenotypic traits in embryo development, neurodevelopment, liver development, and myogenesis, demonstrating that these variations caused a loss of gene function in IARS1. CONCLUSIONS We have found a novel mutation point c.701 T > C (p.L234P) in IARS1. Compound heterozygous mutations of c.701 T > C (p.L234P) and c.1555C > T (p.R519C) in IARS1 are pathogenic, which can cause GRIDHH in child.
Collapse
Affiliation(s)
- Ting-Ting Zou
- Department of Pediatric Infectious Diseases, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hua-Qin Sun
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yu Zhu
- Department of Pediatric Infectious Diseases, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Tian-Tian He
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,Department of Medical Genetics & Prenatal Diagnosis Center, West China Second University Hospital, Sichuan University, No.20, South Section 3, Renmin Road, Chengdu, Sichuan, China
| | - Wen-Wu Ling
- Department of Ultrasound, West China University Hospital, Sichuan University, Chengdu, 610041, China
| | - Hong-Mei Zhu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,Department of Medical Genetics & Prenatal Diagnosis Center, West China Second University Hospital, Sichuan University, No.20, South Section 3, Renmin Road, Chengdu, Sichuan, China
| | - Zi-Yuan Lin
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Yan-Yan Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,Department of Medical Genetics & Prenatal Diagnosis Center, West China Second University Hospital, Sichuan University, No.20, South Section 3, Renmin Road, Chengdu, Sichuan, China
| | - Shan-Ling Liu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,Department of Medical Genetics & Prenatal Diagnosis Center, West China Second University Hospital, Sichuan University, No.20, South Section 3, Renmin Road, Chengdu, Sichuan, China
| | - He Wang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.,SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
| | - Xue-Mei Zhang
- Key Laboratory of Birth Defects and Related Diseases of Women and Children (Sichuan University), Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, China. .,SCU-CUHK Joint Laboratory for Reproductive Medicine, West China Second University Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
34
|
Lehmann V, Schene IF, Ardisasmita AI, Liv N, Veenendaal T, Klumperman J, van der Doef HPJ, Verkade HJ, Verstegen MMA, van der Laan LJW, Jans JJM, Verhoeven‐Duif NM, van Hasselt PM, Nieuwenhuis EES, Spee B, Fuchs SA. The potential and limitations of intrahepatic cholangiocyte organoids to study inborn errors of metabolism. J Inherit Metab Dis 2022; 45:353-365. [PMID: 34671987 PMCID: PMC9298016 DOI: 10.1002/jimd.12450] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 10/11/2021] [Accepted: 10/13/2021] [Indexed: 01/09/2023]
Abstract
Inborn errors of metabolism (IEMs) comprise a diverse group of individually rare monogenic disorders that affect metabolic pathways. Mutations lead to enzymatic deficiency or dysfunction, which results in intermediate metabolite accumulation or deficit leading to disease phenotypes. Currently, treatment options for many IEMs are insufficient. Rarity of individual IEMs hampers therapy development and phenotypic and genetic heterogeneity suggest beneficial effects of personalized approaches. Recently, cultures of patient-own liver-derived intrahepatic cholangiocyte organoids (ICOs) have been established. Since most metabolic genes are expressed in the liver, patient-derived ICOs represent exciting possibilities for in vitro modeling and personalized drug testing for IEMs. However, the exact application range of ICOs remains unclear. To address this, we examined which metabolic pathways can be studied with ICOs and what the potential and limitations of patient-derived ICOs are to model metabolic functions. We present functional assays in patient ICOs with defects in branched-chain amino acid metabolism (methylmalonic acidemia), copper metabolism (Wilson disease), and transporter defects (cystic fibrosis). We discuss the broad range of functional assays that can be applied to ICOs, but also address the limitations of these patient-specific cell models. In doing so, we aim to guide the selection of the appropriate cell model for studies of a specific disease or metabolic process.
Collapse
Affiliation(s)
- Vivian Lehmann
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
- Department of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Imre F. Schene
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Arif I. Ardisasmita
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Nalan Liv
- Section Cell Biology, Center for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Tineke Veenendaal
- Section Cell Biology, Center for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | - Judith Klumperman
- Section Cell Biology, Center for Molecular MedicineUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Henkjan J. Verkade
- Department of Pediatric GastroenterologyUniversity Medical Center GroningenGroningenThe Netherlands
- Department of HepatologyUniversity Medical Center GroningenGroningenThe Netherlands
| | | | | | - Judith J. M. Jans
- Department of Metabolic DiagnosticsUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Peter M. van Hasselt
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| | | | - Bart Spee
- Department of Veterinary MedicineUtrecht UniversityUtrechtThe Netherlands
| | - Sabine A. Fuchs
- Department of Metabolic DiseasesUniversity Medical Center UtrechtUtrechtThe Netherlands
| |
Collapse
|
35
|
Schene IF, Joore IP, Baijens JHL, Stevelink R, Kok G, Shehata S, Ilcken EF, Nieuwenhuis ECM, Bolhuis DP, van Rees RCM, Spelier SA, van der Doef HPJ, Beekman JM, Houwen RHJ, Nieuwenhuis EES, Fuchs SA. Mutation-specific reporter for optimization and enrichment of prime editing. Nat Commun 2022; 13:1028. [PMID: 35232966 PMCID: PMC8888566 DOI: 10.1038/s41467-022-28656-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Accepted: 02/04/2022] [Indexed: 12/23/2022] Open
Abstract
Prime editing is a versatile genome-editing technique that shows great promise for the generation and repair of patient mutations. However, some genomic sites are difficult to edit and optimal design of prime-editing tools remains elusive. Here we present a fluorescent prime editing and enrichment reporter (fluoPEER), which can be tailored to any genomic target site. This system rapidly and faithfully ranks the efficiency of prime edit guide RNAs (pegRNAs) combined with any prime editor variant. We apply fluoPEER to instruct correction of pathogenic variants in patient cells and find that plasmid editing enriches for genomic editing up to 3-fold compared to conventional enrichment strategies. DNA repair and cell cycle-related genes are enriched in the transcriptome of edited cells. Stalling cells in the G1/S boundary increases prime editing efficiency up to 30%. Together, our results show that fluoPEER can be employed for rapid and efficient correction of patient cells, selection of gene-edited cells, and elucidation of cellular mechanisms needed for successful prime editing.
Collapse
Affiliation(s)
- I F Schene
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - I P Joore
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - J H L Baijens
- Utrecht University Graduate School of Life Sciences, Heidelberglaan 8, 3584 CS, Utrecht, The Netherlands
| | - R Stevelink
- Department of Genetics, UMC Utrecht Brain Center, University Medical Center Utrecht, Utrecht University, Heidelberglaan 100, 3584 CX, Utrecht, The Netherlands
| | - G Kok
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - S Shehata
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - E F Ilcken
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - E C M Nieuwenhuis
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - D P Bolhuis
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - R C M van Rees
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
| | - S A Spelier
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
| | - H P J van der Doef
- Department of Pediatric Gastroenterology, University Medical Center Groningen, Hanzeplein 1, 9713 GZ, Groningen, The Netherlands
| | - J M Beekman
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands
- Department of Pediatric Respiratory Medicine, Wilhelmina Children's Hospital, University Medical Center, Utrecht University, 3584 EA, Utrecht, The Netherlands
| | - R H J Houwen
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
| | - E E S Nieuwenhuis
- Division of Pediatric Gastroenterology, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands
- Department of Sciences, University College Roosevelt, Lange Noordstraat 1, 4331 CB, Middelburg, The Netherlands
| | - S A Fuchs
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Lundlaan 6, 3584 EA, Utrecht, The Netherlands.
- Regenerative Medicine Center Utrecht, Uppsalalaan 8, 3584 CT, Utrecht, The Netherlands.
| |
Collapse
|
36
|
Charbit-Henrion F, Goguyer-Deschaumes R, Borensztajn K, Mirande M, Berthelet J, Rodrigues-Lima F, Khiat A, Frémond ML, Bader-Meunier B, Rodari MM, Seabra L, Rice GI, Legendre M, Drummond D, Berteloot L, Roux CJ, Boddaert N, Drabent P, Molina TJ, Lacaille F, Kossorotoff M, Cerf-Bensussan N, Parlato M, Hadchouel A. Systemic inflammatory syndrome in children with FARSA deficiency. Clin Genet 2022; 101:552-558. [PMID: 35132614 PMCID: PMC9303323 DOI: 10.1111/cge.14120] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 01/13/2022] [Accepted: 01/30/2022] [Indexed: 11/27/2022]
Abstract
Variants in aminoacyl-tRNA synthetases (ARSs) genes are associated to a broad spectrum of human inherited diseases. Patients with defective PheRS, encoded by FARSA and FARSB, display brain abnormalities, interstitial lung disease and facial dysmorphism. We investigated four children from two unrelated consanguineous families carrying two missense homozygous variants in FARSA with significantly reduced PheRS-mediated aminoacylation activity. In addition to the core ARS-phenotype, all patients showed an inflammatory profile associated with autoimmunity and interferon score, a clinical feature not ascribed to PheRS-deficient patients to date. JAK inhibition improved lung disease in one patient. Our findings expand the genetic and clinical spectrum of FARSA-related disease. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Fabienne Charbit-Henrion
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, Inserm, UMR1163, Paris, France
| | - Roman Goguyer-Deschaumes
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, Inserm, UMR1163, Paris, France
| | - Keren Borensztajn
- Sorbonne Université, Inserm, Childhood genetic disorders, Hôpital Trousseau, Paris, France
| | - Marc Mirande
- Laboratoire de Biologie et Pharmacologie Appliquée, UMR8113 CNRS, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | | | - Fernando Rodrigues-Lima
- Université de Paris, Unité de Biologie Fonctionnelle et Adaptative, CNRS UMR 8251, Paris, France
| | - Anis Khiat
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, Inserm, UMR1163, Paris, France
| | - Marie-Louise Frémond
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris, France
| | - Brigitte Bader-Meunier
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service d'immuno-hématologie pédiatrique, Paris, France
| | - Marco M Rodari
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, Inserm, UMR1163, Paris, France
| | - Luis Seabra
- Université de Paris, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, Imagine Institute, Paris, France
| | - Gillian I Rice
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| | - Marie Legendre
- Laboratoire de Biologie et Pharmacologie Appliquée, UMR8113 CNRS, ENS Paris-Saclay, Université Paris-Saclay, Gif-sur-Yvette, France
| | - David Drummond
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Paris, France
| | - Laureline Berteloot
- APHP, Hôpital Universitaire Necker-Enfants Malades, Service d'imagerie pédiatrique Paris, France
| | - Charles-Joris Roux
- APHP, Hôpital Universitaire Necker-Enfants Malades, Service d'imagerie pédiatrique Paris, France
| | - Nathalie Boddaert
- APHP, Hôpital Universitaire Necker-Enfants Malades, Service d'imagerie pédiatrique Paris, France
| | - Philippe Drabent
- APHP, Hôpital Universitaire Necker-Enfants Malades, Service d'anatomopathologie, Paris, France
| | - Thierry Jo Molina
- APHP, Hôpital Universitaire Necker-Enfants Malades, Service d'anatomopathologie, Paris, France
| | - Florence Lacaille
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de gastroentérologie et hépatologie pédiatriques, Paris, France
| | - Manoelle Kossorotoff
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Neurologie Pédiatrique, Paris, France
| | - Nadine Cerf-Bensussan
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, Inserm, UMR1163, Paris, France
| | - Marianna Parlato
- Université de Paris, Imagine Institute, Laboratory of Intestinal Immunity, Inserm, UMR1163, Paris, France
| | - Alice Hadchouel
- Division of Evolution and Genomic Sciences, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester Academic Health Science Centre, Manchester, UK
| |
Collapse
|
37
|
Chan DL, Rudinger-Thirion J, Frugier M, Riley LG, Ho G, Kothur K, Mohammad SS. A case of QARS1 associated epileptic encephalopathy and review of epilepsy in aminoacyl-tRNA synthetase disorders. Brain Dev 2022; 44:142-147. [PMID: 34774383 DOI: 10.1016/j.braindev.2021.10.009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Revised: 09/21/2021] [Accepted: 10/22/2021] [Indexed: 12/30/2022]
Abstract
INTRODUCTION Mutations in QARS1, which encodes human glutaminyl-tRNA synthetase, have been associated with epilepsy, developmental regression, progressive microcephaly and cerebral atrophy. Epilepsy caused by variants in QARS1 is usually drug-resistant and intractable. Childhood onset epilepsy is also reported in various aminoacyl-tRNA synthetase disorders. We describe a case with a milder neurological phenotype than previously reported with QARS1 variants and review the seizure associations with aminoacyl-tRNA synthetase disorders. CASE REPORT The patient is a 4-year-old girl presenting at 6 weeks of age with orofacial dyskinesia and hand stereotypies. She developed focal seizures at 7 months of age. Serial electroencephalograms showed shifting focality. Her seizures were controlled after introduction of carbamazepine. Progress MRI showed very mild cortical volume loss without myelination abnormalities or cerebellar atrophy. She was found to have novel compound heterozygous variants in QARS1 (NM_005051.2): c.[1132C > T];[1574G > A], p.[(Arg378Cys)];[(Arg525Gln)] originally classified as "variants of uncertain significance" and later upgraded to "likely pathogenic" based on functional testing and updated variant database review. Functional testing showed reduced solubility of the corresponding QARS1 mutants in vitro, but only mild two-fold loss in catalytic efficiency with the c.1132C > T variant and no noted change in tRNAGln aminoacylation with the c.1574G > A variant. CONCLUSION We describe two QARS1 variants associated with overall conserved tRNA aminoacylation activity but characterized by significantly reduced QARS protein solubility, resulting in a milder clinical phenotype. 86% of previous patients reported with QARS1 had epilepsy and 79% were pharmaco-resistant. We also summarise literature regarding epilepsy in aminoacyl-tRNA synthetase disorders, which is also often early onset, severe and drug-refractory.
Collapse
Affiliation(s)
- Denise L Chan
- Neurology Department, Sydney Children's Hospital, Sydney, Australia, School of Women's and Children's Health, UNSW Medicine, UNSW Sydney, Australia
| | - Joëlle Rudinger-Thirion
- Université de Strasbourg, CNRS,Architecture et Réactivité de l'ARN, UPR 9002, F-67000, Strasbourg, France
| | - Magali Frugier
- Université de Strasbourg, CNRS,Architecture et Réactivité de l'ARN, UPR 9002, F-67000, Strasbourg, France
| | - Lisa G Riley
- Rare Diseases Functional Genomics, Kids Research, Sydney Children's Hospital Network & Children's Medical Research Institute, Sydney Children's Hospital Network, Sydney, NSW 2145, Australia, Discipline of Child & Adolescent Health, Sydney Medical School, University of Sydney, Sydney, NSW 2006, Australia
| | - Gladys Ho
- Sydney Genome Diagnostics, Western Sydney Genetics Program, The Children's Hospital at Westmead, Sydney, Australia, Discipline of Child & Adolescent Health, Discipline of Genetic Medicine, The University of Sydney, Sydney, Australia; Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Kavitha Kothur
- Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia
| | - Shekeeb S Mohammad
- Children's Hospital at Westmead Clinical School, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW, Australia; TY Nelson Department of Neurology and Neurosurgery, The Children's Hospital at Westmead, Sydney, NSW, Australia, Kids Neuroscience Centre, Kids Research, The Children's Hospital at Westmead, The University of Sydney, Sydney, NSW, Australia.
| |
Collapse
|
38
|
Giong HK, Lee JS. Systematic expression profiling of neuropathy-related aminoacyl-tRNA synthetases in zebrafish during development. Biochem Biophys Res Commun 2022; 587:92-98. [PMID: 34872004 DOI: 10.1016/j.bbrc.2021.11.098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2021] [Accepted: 11/27/2021] [Indexed: 12/01/2022]
Abstract
Aminoacyl tRNA synthetases (ARSs) are a group of proteins, acting as transporters to transfer and attach the appropriate amino acids onto their cognate tRNAs for translation. So far, 18 out of 20 cytoplasmic ARSs are reported to be connected to different neuropathy disorders with multi-organ defects that are often accompanied with developmental delays. Thus, it is important to understand functions and impacts of ARSs at the whole organism level. Here, we systematically analyzed the spatiotemporal expression of 14 ars and 2 aimp genes during development in zebrafish that have not be previously reported. Not only in the brain, their dynamic expression patterns in several tissues such as in the muscles, liver and intestine suggest diverse roles in a wide range of development processes in addition to neuronal function, which is consistent with potential involvement in multiple syndrome diseases associated with ARS mutations. In particular, hinted by its robust expression pattern in the brain, we confirmed that aimp1 is required for the formation of cerebrovasculature by a loss-of-function approach. Overall, our systematic profiling data provides a useful basis for studying roles of ARSs during development and understanding their potential functions in the etiology of related diseases.
Collapse
Affiliation(s)
- Hoi-Khoanh Giong
- Disease Target Structure Research Center, KRIBB, Daejeon, South Korea; KRIBB School, University of Science and Technology, Daejeon, South Korea; Dementia DTC R&D Convergence Program, KIST, Seoul, South Korea
| | - Jeong-Soo Lee
- Disease Target Structure Research Center, KRIBB, Daejeon, South Korea; KRIBB School, University of Science and Technology, Daejeon, South Korea; Dementia DTC R&D Convergence Program, KIST, Seoul, South Korea.
| |
Collapse
|
39
|
O'Callaghan M, Helly F, Tarling E, Keane MP, McCarthy C. Methionine supplementation; potential for improving alveolar macrophage function through reverse cholesterol transport? Eur Respir J 2021; 59:13993003.02594-2021. [PMID: 34857611 DOI: 10.1183/13993003.02594-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Accepted: 10/25/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Marissa O'Callaghan
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Feargal Helly
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland
| | - Elizabeth Tarling
- Division of Cardiology, University of California, Los Angeles, California, USA
| | - Michael P Keane
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland.,School of Medicine, University College Dublin, Dublin, Ireland
| | - Cormac McCarthy
- Department of Respiratory Medicine, St. Vincent's University Hospital, Dublin, Ireland .,School of Medicine, University College Dublin, Dublin, Ireland
| |
Collapse
|
40
|
WARS2 mutations cause dopa-responsive early-onset parkinsonism and progressive myoclonus ataxia. Parkinsonism Relat Disord 2021; 94:54-61. [PMID: 34890876 DOI: 10.1016/j.parkreldis.2021.11.030] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 10/12/2021] [Accepted: 11/28/2021] [Indexed: 11/22/2022]
Abstract
INTRODUCTION Sixteen subjects with biallelic WARS2 variants encoding the tryptophanyl mitochondrial aminoacyl-tRNA synthetase, presenting with a neonatal- or infantile-onset mitochondrial disease, have been reported to date. Here we present six novel cases with WARS2-related diseases and expand the spectrum to later onset phenotypes including dopa-responsive early-onset parkinsonism and progressive myoclonus-ataxia. METHODS Six individuals from four families underwent whole-exome sequencing within research and diagnostic settings. Following the identification of a genetic defect, in-depth phenotyping and protein expression studies were performed. RESULTS A relatively common (gnomAD MAF = 0.0033) pathogenic p.(Trp13Gly) missense variant in WARS2 was detected in trans in all six affected individuals in combination with different pathogenic alleles (exon 2 deletion in family 1; p.(Leu100del) in family 2; p.(Gly50Asp) in family 3; and p.(Glu208*) in family 4). Two subjects presented with action tremor around age 10-12 years and developed tremor-dominant parkinsonism with prominent neuropsychiatric features later in their 20s. Two subjects presented with a progressive myoclonus-ataxia dominant phenotype. One subject presented with spasticity, choreo-dystonia, myoclonus, and speech problems. One subject presented with speech problems, ataxia, and tremor. Western blotting analyses in patient-derived fibroblasts showed a markedly decreased expression of the full-length WARS2 protein in both subjects carrying p.(Trp13Gly) and an exon-2 deletion in compound heterozygosity. CONCLUSIONS This study expands the spectrum of the disease to later onset phenotypes of early-onset tremor-dominant parkinsonism and progressive myoclonus-ataxia phenotypes.
Collapse
|
41
|
Hadchouel A, Delacourt C. Comment on: Methionine supplementation; potential for improving alveolar macrophage function through reverse cholesterol transport? Eur Respir J 2021; 59:13993003.02937-2021. [PMID: 34857610 DOI: 10.1183/13993003.02937-2021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 11/24/2021] [Indexed: 11/05/2022]
Affiliation(s)
- Alice Hadchouel
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France .,Faculté de Médecine, Université de Paris, Paris, France
| | - Christophe Delacourt
- AP-HP, Hôpital Universitaire Necker-Enfants Malades, Service de Pneumologie Pédiatrique, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France
| |
Collapse
|
42
|
Averdunk L, Sticht H, Surowy H, Lüdecke HJ, Koch-Hogrebe M, Alsaif HS, Kahrizi K, Alzaidan H, Alawam BS, Tohary M, Kraus C, Endele S, Wadman E, Kaplan JD, Efthymiou S, Najmabadi H, Reis A, Alkuraya FS, Wieczorek D. The recurrent missense mutation p.(Arg367Trp) in YARS1 causes a distinct neurodevelopmental phenotype. J Mol Med (Berl) 2021; 99:1755-1768. [PMID: 34536092 PMCID: PMC8599376 DOI: 10.1007/s00109-021-02124-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 07/21/2021] [Accepted: 07/30/2021] [Indexed: 11/14/2022]
Abstract
Pathogenic variants in aminoacyl-tRNA synthetases (ARS1) cause a diverse spectrum of autosomal recessive disorders. Tyrosyl tRNA synthetase (TyrRS) is encoded by YARS1 (cytosolic, OMIM*603,623) and is responsible of coupling tyrosine to its specific tRNA. Next to the enzymatic domain, TyrRS has two additional functional domains (N-Terminal TyrRSMini and C-terminal EMAP-II-like domain) which confer cytokine-like functions. Mutations in YARS1 have been associated with autosomal-dominant Charcot-Marie-Tooth (CMT) neuropathy type C and a heterogenous group of autosomal recessive, multisystem diseases. We identified 12 individuals from 6 families with the recurrent homozygous missense variant c.1099C > T;p.(Arg367Trp) (NM_003680.3) in YARS1. This variant causes a multisystem disorder with developmental delay, microcephaly, failure to thrive, short stature, muscular hypotonia, ataxia, brain anomalies, microcytic anemia, hepatomegaly, and hypothyroidism. In silico analyses show that the p.(Arg367Trp) does not affect the catalytic domain responsible of enzymatic coupling, but destabilizes the cytokine-like C-terminal domain. The phenotype associated with p.(Arg367Trp) is distinct from the other biallelic pathogenic variants that reside in different functional domains of TyrRS which all show some common, but also divergent clinical signs [(e.g., p.(Phe269Ser)-retinal anomalies, p.(Pro213Leu)/p.(Gly525Arg)-mild ID, p.(Pro167Thr)-high fatality)]. The diverse clinical spectrum of ARS1-associated disorders is related to mutations affecting the various non-canonical domains of ARS1, and impaired protein translation is likely not the exclusive disease-causing mechanism of YARS1- and ARS1-associated neurodevelopmental disorders. KEY MESSAGES: The missense variant p.(Arg367Trp) in YARS1 causes a distinct multisystem disorder. p.(Arg367Trp) affects a non-canonical domain with cytokine-like functions. Phenotypic heterogeneity associates with the different affected YARS1 domains. Impaired protein translation is likely not the exclusive mechanism of ARS1-associated disorders.
Collapse
Affiliation(s)
- Luisa Averdunk
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
- Department of General Pediatrics, Neonatology and Pediatric Cardiology, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University, Düsseldorf, Germany
| | - Heinrich Sticht
- Division of Bioinformatics, Institute of Biochemistry, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Harald Surowy
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | - Hermann-Josef Lüdecke
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany
| | | | - Hessa S Alsaif
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Kimia Kahrizi
- Genetics Research Center, University of Social Welfare and Rehabilitation Sciences, Tehran, Iran
| | - Hamad Alzaidan
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Bashayer S Alawam
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Mohamed Tohary
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Cornelia Kraus
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Sabine Endele
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Erin Wadman
- Division of Medical Genetics, Department of Pediatrics, Nemours Alfred I, DuPont Hospital for Children, Wilmington, Delaware, DE, USA
| | - Julie D Kaplan
- Division of Medical Genetics, Department of Pediatrics, Nemours Alfred I, DuPont Hospital for Children, Wilmington, Delaware, DE, USA
| | - Stephanie Efthymiou
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology and The National Hospital for Neurology and Neurosurgery, London, UK
| | - Hossein Najmabadi
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - André Reis
- Institute of Human Genetics, University Hospital Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Dagmar Wieczorek
- Institute of Human Genetics, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine-University Düsseldorf, Universitätsstraße 1, 40225, Düsseldorf, Germany.
| |
Collapse
|
43
|
Kok G, van Karnebeek CDM, Fuchs SA. Response to Shen et al. Genet Med 2021; 24:506-507. [PMID: 34906483 DOI: 10.1016/j.gim.2021.09.022] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 09/29/2021] [Accepted: 09/30/2021] [Indexed: 11/28/2022] Open
Affiliation(s)
- Gautam Kok
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Clara D M van Karnebeek
- Department of Pediatrics & Metabolic Diseases, Radboud Centre for Mitochondrial Medicine, Amalia Children's Hospital, Radboud University Medical Center, Nijmegen, The Netherlands; Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Centers, Amsterdam, The Netherlands
| | - Sabine A Fuchs
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands.
| |
Collapse
|
44
|
Lin SJ, Vona B, Barbalho PG, Kaiyrzhanov R, Maroofian R, Petree C, Severino M, Stanley V, Varshney P, Bahena P, Alzahrani F, Alhashem A, Pagnamenta AT, Aubertin G, Estrada-Veras JI, Hernández HAD, Mazaheri N, Oza A, Thies J, Renaud DL, Dugad S, McEvoy J, Sultan T, Pais LS, Tabarki B, Villalobos-Ramirez D, Rad A, Galehdari H, Ashrafzadeh F, Sahebzamani A, Saeidi K, Torti E, Elloumi HZ, Mora S, Palculict TB, Yang H, Wren JD, Ben Fowler, Joshi M, Behra M, Burgess SM, Nath SK, Hanna MG, Kenna M, Merritt JL, Houlden H, Karimiani EG, Zaki MS, Haaf T, Alkuraya FS, Gleeson JG, Varshney GK. Biallelic variants in KARS1 are associated with neurodevelopmental disorders and hearing loss recapitulated by the knockout zebrafish. Genet Med 2021; 23:1933-1943. [PMID: 34172899 PMCID: PMC8956360 DOI: 10.1038/s41436-021-01239-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 12/04/2022] Open
Abstract
PURPOSE Pathogenic variants in Lysyl-tRNA synthetase 1 (KARS1) have increasingly been recognized as a cause of early-onset complex neurological phenotypes. To advance the timely diagnosis of KARS1-related disorders, we sought to delineate its phenotype and generate a disease model to understand its function in vivo. METHODS Through international collaboration, we identified 22 affected individuals from 16 unrelated families harboring biallelic likely pathogenic or pathogenic in KARS1 variants. Sequencing approaches ranged from disease-specific panels to genome sequencing. We generated loss-of-function alleles in zebrafish. RESULTS We identify ten new and four known biallelic missense variants in KARS1 presenting with a moderate-to-severe developmental delay, progressive neurological and neurosensory abnormalities, and variable white matter involvement. We describe novel KARS1-associated signs such as autism, hyperactive behavior, pontine hypoplasia, and cerebellar atrophy with prevalent vermian involvement. Loss of kars1 leads to upregulation of p53, tissue-specific apoptosis, and downregulation of neurodevelopmental related genes, recapitulating key tissue-specific disease phenotypes of patients. Inhibition of p53 rescued several defects of kars1-/- knockouts. CONCLUSION Our work delineates the clinical spectrum associated with KARS1 defects and provides a novel animal model for KARS1-related human diseases revealing p53 signaling components as potential therapeutic targets.
Collapse
Affiliation(s)
- Sheng-Jia Lin
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Barbara Vona
- Department of Otolaryngology-Head & Neck Surgery, Tübingen Hearing Research Centre, Eberhard Karls University of Tübingen, Tübingen, Germany.,Institute of Human Genetics, Julius Maximilians University Würzburg, Würzburg, Germany
| | - Patricia G Barbalho
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Rauan Kaiyrzhanov
- Department of Neuromuscular Disorders, Queen Square Institute of Neurology, University College London, London, UK
| | - Reza Maroofian
- Department of Neuromuscular Disorders, Queen Square Institute of Neurology, University College London, London, UK
| | - Cassidy Petree
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | | | - Valentina Stanley
- Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Pratishtha Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Paulina Bahena
- Institute of Human Genetics, Julius Maximilians University Würzburg, Würzburg, Germany
| | - Fatema Alzahrani
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia
| | - Amal Alhashem
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Alistair T Pagnamenta
- NIHR Biomedical Research Centre, Wellcome Centre for Human Genetics, University of Oxford, Oxford, UK
| | - Gudrun Aubertin
- Division of Medical Genetics, Department of Pathology and Lab Medicine, Island Health, Victoria General Hospital, Victoria, BC, Canada
| | - Juvianee I Estrada-Veras
- Henry M Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA.,Pediatric Subspecialty Genetics Walter Reed National Military Medical Center, Bethesda, MD, USA.,Murtha Cancer Center / Research Program, Department of Surgery, Uniformed Services University of the Health Sciences, Bethesda, MD, USA
| | - Héctor Adrián Díaz Hernández
- Department of Gastrointestinal Endoscopy, National Institute of Medical Sciences and Nutrition Salvador Zubirán, Mexico City, Mexico
| | - Neda Mazaheri
- Department of Genetics, Faculty of Science, Shahid Chamran University of Ahvaz, Ahvaz, Iran.,Narges Medical Genetics and Prenatal Diagnostics Laboratory, East Mihan Ave., Kianpars, Iran
| | - Andrea Oza
- Otolaryngology and Communication Enhancement, Boston Children's Hospital, and Dept. of Otolaryngology, Harvard medical School, Boston, USA
| | - Jenny Thies
- Department of Biochemical Genetics, Seattle Children's Hospital, Seattle, WA, USA
| | - Deborah L Renaud
- Departments of Neurology and Pediatrics, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
| | - Sanmati Dugad
- Bioinformatics Centre, S. P. Pune University, Pune, India
| | - Jennifer McEvoy
- Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Tipu Sultan
- Department of Pediatric Neurology, Children's Hospital and Institute of Child Health, Lahore, Pakistan
| | - Lynn S Pais
- Broad Center for Mendelian Genomics, Program in Medical and Population Genetics, Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, MA, USA
| | - Brahim Tabarki
- Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | | | - Aboulfazl Rad
- Department of Otolaryngology-Head & Neck Surgery, Tübingen Hearing Research Centre, Eberhard Karls University of Tübingen, Tübingen, Germany
| | | | - Hamid Galehdari
- Department of Gastrointestinal Endoscopy, National Institute of Medical Sciences and Nutrition Salvador Zubirán, Mexico City, Mexico
| | - Farah Ashrafzadeh
- Department of Pediatric Diseases, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Afsaneh Sahebzamani
- Pediatric and Genetic Counselling Center, Kerman Welfare Organization, Kerman, Iran
| | - Kolsoum Saeidi
- Neuroscience Research Center, Institute of Neuropharmacology, Kerman University of Medical Sciences, Kerman, Iran
| | - Erin Torti
- GeneDx, 207 Perry Parkway Gaithersburg, Gaithersburg, MD, USA
| | - Houda Z Elloumi
- GeneDx, 207 Perry Parkway Gaithersburg, Gaithersburg, MD, USA
| | - Sara Mora
- GeneDx, 207 Perry Parkway Gaithersburg, Gaithersburg, MD, USA
| | | | - Hui Yang
- GeneDx, 207 Perry Parkway Gaithersburg, Gaithersburg, MD, USA
| | - Jonathan D Wren
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Ben Fowler
- Imaging core facility, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Manali Joshi
- Bioinformatics Centre, S. P. Pune University, Pune, India
| | - Martine Behra
- Department of Neurobiology, University of Puerto Rico, San Juan, PR, USA
| | - Shawn M Burgess
- Translational & Functional Genomics Branch, National Human Genome Research Institute, NIH, Bethesda, MD, USA
| | - Swapan K Nath
- Arthritis & Clinical Immunology Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
| | - Michael G Hanna
- Department of Neuromuscular Disorders, Queen Square Institute of Neurology, University College London, London, UK
| | - Margaret Kenna
- Otolaryngology and Communication Enhancement, Boston Children's Hospital, and Dept. of Otolaryngology, Harvard medical School, Boston, USA
| | - J Lawrence Merritt
- Department of Pediatrics, Biochemical Genetics, University of Washington, Seattle, WA, USA
| | - Henry Houlden
- Department of Neuromuscular Disorders, Queen Square Institute of Neurology, University College London, London, UK
| | - Ehsan Ghayoor Karimiani
- Molecular and Clinical Sciences Institute, St. George's, University of London, Cranmer Terrace London, London, UK.,Innovative Medical Research Center, Mashhad Branch, Islamic Azdad University, Mashhad, Iran
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo, Egypt
| | - Thomas Haaf
- Institute of Human Genetics, Julius Maximilians University Würzburg, Würzburg, Germany
| | - Fowzan S Alkuraya
- Department of Genetics, King Faisal Specialist Hospital and Research Center, Riyadh, Saudi Arabia.,Department of Pediatrics, Prince Sultan Military Medical City, Riyadh, Saudi Arabia
| | - Joseph G Gleeson
- Department of Neurosciences, Rady Children's Institute for Genomic Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gaurav K Varshney
- Genes & Human Disease Research Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, USA.
| |
Collapse
|
45
|
Ravel JM, Dreumont N, Mosca P, Smith DEC, Mendes MI, Wiedemann A, Coelho D, Schmitt E, Rivière JB, Tran Mau-Them F, Thevenon J, Kuentz P, Polivka M, Fuchs SA, Kok G, Thauvin-Robinet C, Guéant JL, Salomons GS, Faivre L, Feillet F. A bi-allelic loss-of-function SARS1 variant in children with neurodevelopmental delay, deafness, cardiomyopathy, and decompensation during fever. Hum Mutat 2021; 42:1576-1583. [PMID: 34570399 DOI: 10.1002/humu.24285] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 09/01/2021] [Accepted: 09/23/2021] [Indexed: 11/08/2022]
Abstract
Aminoacyl-tRNA synthetases (aaRS) are ubiquitously expressed enzymes responsible for ligating amino acids to their cognate tRNA molecules through an aminoacylation reaction. The resulting aminoacyl-tRNA is delivered to ribosome elongation factors to participate in protein synthesis. Seryl-tRNA synthetase (SARS1) is one of the cytosolic aaRSs and catalyzes serine attachment to tRNASer . SARS1 deficiency has already been associated with moderate intellectual disability, ataxia, muscle weakness, and seizure in one family. We describe here a new clinical presentation including developmental delay, central deafness, cardiomyopathy, and metabolic decompensation during fever leading to death, in a consanguineous Turkish family, with biallelic variants (c.638G>T, p.(Arg213Leu)) in SARS1. This missense variant was shown to lead to protein instability, resulting in reduced protein level and enzymatic activity. Our results describe a new clinical entity and expand the clinical and mutational spectrum of SARS1 and aaRS deficiencies.
Collapse
Affiliation(s)
- Jean-Marie Ravel
- Reference Centre of Inborn Metabolism Diseases, Université de Lorraine, CHRU-Nancy, Nancy, France.,NGERE, Université de Lorraine, Inserm, Nancy, France
| | | | - Pauline Mosca
- NGERE, Université de Lorraine, Inserm, Nancy, France
| | - Desiree E C Smith
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Marisa I Mendes
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | | | - David Coelho
- NGERE, Université de Lorraine, Inserm, Nancy, France
| | | | - Jean-Baptiste Rivière
- Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France.,Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Compétence Maladies Mitochondriales, FHU TRANSLAD, Hôpital d'Enfants, CHU de Dijon, France.,INSERM UMR1231, Equipe Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Frédéric Tran Mau-Them
- Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France.,Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Compétence Maladies Mitochondriales, FHU TRANSLAD, Hôpital d'Enfants, CHU de Dijon, France.,INSERM UMR1231, Equipe Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Julien Thevenon
- Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France.,Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Compétence Maladies Mitochondriales, FHU TRANSLAD, Hôpital d'Enfants, CHU de Dijon, France.,INSERM UMR1231, Equipe Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Paul Kuentz
- Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Compétence Maladies Mitochondriales, FHU TRANSLAD, Hôpital d'Enfants, CHU de Dijon, France.,INSERM UMR1231, Equipe Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Marc Polivka
- Department of Pathology, Hôpital Lariboisière, Paris, France
| | - Sabine A Fuchs
- Department of Metabolic Diseases, Wilhelmina Children's Hospital, University Medical Center Utrecht, Utrecht, The Netherlands.,Regenerative Medicine Center Utrecht, Regenerative Medicine Utrecht, Utrecht, The Netherlands.,On behalf of "United for Metabolic Diseases,", Amsterdam, the Netherlands
| | - Gautam Kok
- Department of Pathology, Hôpital Lariboisière, Paris, France
| | - Christel Thauvin-Robinet
- Unité Fonctionnelle Innovation en Diagnostic génomique des maladies rares, CHU Dijon Bourgogne, Dijon, France.,Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Compétence Maladies Mitochondriales, FHU TRANSLAD, Hôpital d'Enfants, CHU de Dijon, France.,INSERM UMR1231, Equipe Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - Jean-Louis Guéant
- Reference Centre of Inborn Metabolism Diseases, Université de Lorraine, CHRU-Nancy, Nancy, France.,NGERE, Université de Lorraine, Inserm, Nancy, France
| | - Gajja S Salomons
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Laurence Faivre
- Centre de Génétique, Centre de Référence Anomalies du Développement et Syndromes Malformatifs, Centre de Compétence Maladies Mitochondriales, FHU TRANSLAD, Hôpital d'Enfants, CHU de Dijon, France.,INSERM UMR1231, Equipe Génétique des Anomalies du Développement, Université de Bourgogne, Dijon, France
| | - François Feillet
- Reference Centre of Inborn Metabolism Diseases, Université de Lorraine, CHRU-Nancy, Nancy, France.,NGERE, Université de Lorraine, Inserm, Nancy, France
| |
Collapse
|
46
|
La Fay C, Hoebeke C, Juzaud M, Spraul A, Heux P, Dubus JC, Hadchouel A, Fabre A. Deep phenotyping of MARS1 (interstitial lung and liver disease) and LARS1 (infantile liver failure syndrome 1) recessive multisystemic disease using Human Phenotype Ontology annotation: Overlap and differences. Case report and review of literature. Eur J Med Genet 2021; 64:104334. [PMID: 34496286 DOI: 10.1016/j.ejmg.2021.104334] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 09/02/2021] [Indexed: 02/07/2023]
Abstract
INTRODUCTION Aminoacyl transfer RNA (tRNA) synthetases are associated with diseases when mutations occur in their encoding genes. Pulmonary alveolar proteinosis can be caused by mutation in the methionyl-tRNA synthetase (MARS) gene while mutations in the leucine-tRNA synthetase (LARS) gene lead to infantile liver failure syndrome type 1. We report the case of a patient with LARS1 pathogenics variants and two patients with MARS1 pathogenics variants. The aim of this study was to analyze the phenotypes of our three patients in detail and classify cases in the literature using Human Phenotype Ontology (HPO) terms. RESULTS The first patient has two previously undescribed heterozygous variants in LARS1 (c.1818dup and c.463A>G). The other two patients' MARS1 variants (c.1177G>A and c.1700C>T) have already been described in the literature. All three patients had anemia, hepatomegaly, feeding difficulties, failure to thrive and hypoalbuminemia. Including ours, 65 patients are described in total, for whom 117 phenotypic abnormalities have been described at least once, 41.9% of which both in patients with LARS1 and MARS1 mutations. CONCLUSION Patients with LARS1 and MARS1 mutations seem to share a common phenotype but further deep phenotyping studies are required to clarify the details of these complex pathologies.
Collapse
Affiliation(s)
- Charlotte La Fay
- Department of Pediatric Gastroenterology and Hepatology, Multidisciplinary Pediatric, Aix Marseille University, La Timone Children Hospital, AP-HM, 13005 Marseille, France.
| | - Celia Hoebeke
- Department of Neuropediatrics and Metabolism, Reference Center of Inherited Metabolic Disorders, La Timone Children Hospital, Marseille, France
| | - Marine Juzaud
- Department of Pediatric Gastroenterology and Hepatology, Multidisciplinary Pediatric, Aix Marseille University, La Timone Children Hospital, AP-HM, 13005 Marseille, France
| | - Anne Spraul
- AP-HP, Hospital Bicêtre, DMU15, Service de Biochimie, Le Kremlin Bicêtre, France
| | - Pauline Heux
- Aix Marseille University, INSERM, MMG, Marseille, France
| | - Jean-Christophe Dubus
- Service de Médecine Infantile et Pneumologie Pédiatrique, CHU Timone-Enfants, 264 Rue Saint-Pierre, 13385, Marseille Cedex 5, France; Aix-Marseille Université, IRD, AP-HM, MEPHI, IHU Méditerranée-Infection France
| | - Alice Hadchouel
- Pediatric Pulmonology, AP-HP, University Hospital Necker-Enfants Malades, Paris, France
| | - Alexandre Fabre
- Pediatric Multidisciplinary Pediatric APHM, Timone Enfant, Marseille, France; Aix-Marseille University, INSERM, GMGF, Marseille, France
| |
Collapse
|
47
|
Hadchouel A, Drummond D, Pontoizeau C, Aoust L, Hurtado Nedelec MM, El Benna J, Gachelin E, Perisson C, Vigier C, Schiff M, Lacaille F, Molina TJ, Berteloot L, Renolleau S, Ottolenghi C, Tréluyer JM, de Blic J, Delacourt C. Methionine supplementation for multi-organ dysfunction in MetRS-related pulmonary alveolar proteinosis. Eur Respir J 2021; 59:13993003.01554-2021. [PMID: 34503986 DOI: 10.1183/13993003.01554-2021] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Accepted: 08/17/2021] [Indexed: 11/09/2022]
Abstract
INTRODUCTION Pulmonary alveolar proteinosis related to mutations in the methionine tRNA synthetase (MARS1) gene is a severe, early-onset disease that results in death before the age of 2 years in one-third of patients. It is associated with a liver disease, growth failure and systemic inflammation. As methionine supplementation in yeast models restored normal enzymatic activity of the synthetase, we studied the tolerance, safety and efficacy of daily oral methionine supplementation in patients with severe and early disease. METHODS Four patients received methionine supplementation and were followed for respiratory, hepatic, growth, and inflammation-related outcomes. Their course was compared to those of historical controls. Reactive oxygen species (ROS) production by patient monocytes before and after methionine supplementation was also studied. RESULTS Methionine supplementation was associated with respiratory improvement, clearance of the extracellular lipoproteinaceous material, and discontinuation of whole-lung lavage in all patients. The three patients who required oxygen or non-invasive ventilation could be weaned off within 60 days. Liver dysfunction, inflammation, and growth delay also improved or resolved. At a cellular level, methionine supplementation normalised the production of reactive oxygen species by peripheral monocytes. CONCLUSION Methionine supplementation was associated with important improvements in children with pulmonary alveolar proteinosis related to mutations in the MARS1 gene. This study paves the way for similar strategies for other tRNA synthetase deficiencies.
Collapse
Affiliation(s)
- Alice Hadchouel
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France .,Faculté de Médecine, Université de Paris, Paris, France
| | - David Drummond
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France
| | - Clément Pontoizeau
- Faculté de Médecine, Université de Paris, Paris, France.,AP-HP, UF de Métabolomique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Laura Aoust
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France
| | - Maria-Margarita Hurtado Nedelec
- INSERM-U1149, Faculté de Médecine, Centre de Recherche sur l'Inflammation (CRI), CNRS-ERL8252, Laboratoire d'Excellence Inflamex, Université Paris Diderot-Sorbonne Paris Cité, , Paris, France.,AP-HP, UF Dysfonctionnements Immunitaires, Centre Hospitalier Universitaire Xavier Bichat, Paris, France
| | - Jamel El Benna
- INSERM-U1149, Faculté de Médecine, Centre de Recherche sur l'Inflammation (CRI), CNRS-ERL8252, Laboratoire d'Excellence Inflamex, Université Paris Diderot-Sorbonne Paris Cité, , Paris, France
| | - Elsa Gachelin
- Service de Pédiatrie, CHU Reunion site Félix Guyon, Saint Denis, France
| | | | | | - Manuel Schiff
- AP-HP, Service de Maladies Héréditaires du Métabolisme, Hôpital Necker-Enfants Malades, Centre de Référence Maladies Héréditaires du Métabolisme, Paris, France.,Institut Imagine, Inserm UMRS 1163, Paris, France
| | - Florence Lacaille
- AP-HP, Service de Gastroentérologie-Hépatologie-Nutrition Pédiatrique, Hôpital Necker-Enfants Malades, Paris, France
| | - Thierry Jo Molina
- Institut Imagine, Inserm UMRS 1163, Paris, France.,AP-HP, Service de Pathologie, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Laureline Berteloot
- Institut Imagine, Inserm UMRS 1163, Paris, France.,AP-HP, Service d'Imagerie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Sylvain Renolleau
- Faculté de Médecine, Université de Paris, Paris, France.,AP-HP, Service de Réanimation médico-chirurgicale pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Chris Ottolenghi
- Faculté de Médecine, Université de Paris, Paris, France.,AP-HP, UF de Métabolomique, Hôpital Universitaire Necker-Enfants Malades, Paris, France
| | - Jean-Marc Tréluyer
- Faculté de Médecine, Université de Paris, Paris, France.,Groupe Hospitalier APHP Centre Université de Paris Recherche Clinique et Pharmacologie Necker Cochin, Paris, France
| | - Jacques de Blic
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France.,Authors contributed equally to this article
| | - Christophe Delacourt
- AP-HP, Service de Pneumologie Pédiatrique, Hôpital Universitaire Necker-Enfants Malades, Centre de Référence pour les Maladies Respiratoires Rares de l'Enfant, Paris, France.,Faculté de Médecine, Université de Paris, Paris, France.,Authors contributed equally to this article
| |
Collapse
|
48
|
Helman G, Mendes MI, Nicita F, Darbelli L, Sherbini O, Moore T, Derksen A, Amy Pizzino, Carrozzo R, Torraco A, Catteruccia M, Aiello C, Goffrini P, Figuccia S, Smith DEC, Hadzsiev K, Hahn A, Biskup S, Brösse I, Kotzaeridou U, Gauck D, Grebe TA, Elmslie F, Stals K, Gupta R, Bertini E, Thiffault I, Taft RJ, Schiffmann R, Brandl U, Haack TB, Salomons GS, Simons C, Bernard G, van der Knaap MS, Vanderver A, Husain RA. Expanded phenotype of AARS1-related white matter disease. Genet Med 2021; 23:2352-2359. [PMID: 34446925 DOI: 10.1038/s41436-021-01286-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 07/08/2021] [Accepted: 07/09/2021] [Indexed: 11/09/2022] Open
Abstract
PURPOSE Recent reports of individuals with cytoplasmic transfer RNA (tRNA) synthetase-related disorders have identified cases with phenotypic variability from the index presentations. We sought to assess phenotypic variability in individuals with AARS1-related disease. METHODS A cross-sectional survey was performed on individuals with biallelic variants in AARS1. Clinical data, neuroimaging, and genetic testing results were reviewed. Alanyl tRNA synthetase (AlaRS) activity was measured in available fibroblasts. RESULTS We identified 11 affected individuals. Two phenotypic presentations emerged, one with early infantile-onset disease resembling the index cases of AARS1-related epileptic encephalopathy with deficient myelination (n = 7). The second (n = 4) was a later-onset disorder, where disease onset occurred after the first year of life and was characterized on neuroimaging by a progressive posterior predominant leukoencephalopathy evolving to include the frontal white matter. AlaRS activity was significantly reduced in five affected individuals with both early infantile-onset and late-onset phenotypes. CONCLUSION We suggest that variants in AARS1 result in a broader clinical spectrum than previously appreciated. The predominant form results in early infantile-onset disease with epileptic encephalopathy and deficient myelination. However, a subgroup of affected individuals manifests with late-onset disease and similarly rapid progressive clinical decline. Longitudinal imaging and clinical follow-up will be valuable in understanding factors affecting disease progression and outcome.
Collapse
Affiliation(s)
- Guy Helman
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Marisa I Mendes
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Francesco Nicita
- Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Lama Darbelli
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Pediatrics, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Omar Sherbini
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Travis Moore
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Alexa Derksen
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada
| | - Amy Pizzino
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Rosalba Carrozzo
- Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Alessandra Torraco
- Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Michela Catteruccia
- Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Chiara Aiello
- Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Paola Goffrini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Sonia Figuccia
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
| | - Desiree E C Smith
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Kinga Hadzsiev
- Department of Medical Genetics, University of Pécs, Pécs, Hungary
| | - Andreas Hahn
- Department of Child Neurology, Justus-Liebig-University, Giessen, Germany
| | - Saskia Biskup
- Praxis fuer Humangenetik and CeGaT GmbH, Tuebingen, Germany
| | - Ines Brösse
- Division of Child Neurology and Inherited Metabolic Diseases, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Urania Kotzaeridou
- Division of Child Neurology and Inherited Metabolic Diseases, Centre for Paediatrics and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Darja Gauck
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Theresa A Grebe
- Division of Genetics and Metabolism, Department of Child Health, Phoenix Children's Hospital, University of Arizona College of Medicine, Phoenix, AZ, USA
| | - Frances Elmslie
- South West Thames Regional Genetics Service, St George's University Hospital, London, UK
| | - Karen Stals
- Molecular Genetics Department, Royal Devon and Exeter NHS Foundation Trust, Exeter, UK
| | - Rajat Gupta
- Department of Neurology, Birmingham Children's Hospital, Birmingham, UK
| | - Enrico Bertini
- Department of Neurosciences, Unit of Muscular and Neurodegenerative Disorders, Laboratory of Molecular Medicine, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Isabelle Thiffault
- Children's Mercy Kansas City, Center for Pediatric Genomic Medicine, Kansas City, MO, USA.,Department of Pathology and Laboratory Medicine, Children's Mercy Hospitals, Kansas City, MO, USA.,School of Medicine, University of Missouri-Kansas City, Kansas City, MO, USA
| | | | | | - Ulrich Brandl
- Department of Neuropediatrics, Jena University Hospital, Jena, Germany
| | - Tobias B Haack
- Institute of Medical Genetics and Applied Genomics, University of Tuebingen, Tuebingen, Germany
| | - Gajja S Salomons
- Metabolic Unit, Department of Clinical Chemistry, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - Cas Simons
- Murdoch Children's Research Institute, The Royal Children's Hospital, Parkville, VIC, Australia.,Institute for Molecular Bioscience, The University of Queensland, Brisbane, Queensland, Australia
| | - Geneviève Bernard
- Department of Neurology and Neurosurgery, McGill University, Montreal, QC, Canada.,Child Health and Human Development Program, Research Institute of the McGill University Health Centre, Montreal, QC, Canada.,Department of Pediatrics, McGill University, Montreal, QC, Canada.,Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Marjo S van der Knaap
- Department of Child Neurology, Emma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit Amsterdam and Amsterdam Neuroscience, Amsterdam, The Netherlands.,Department of Functional Genomics, Center for Neurogenomics and Cognitive Research, VU University, Amsterdam, The Netherlands
| | - Adeline Vanderver
- Division of Neurology, Children's Hospital of Philadelphia, Philadelphia, PA, USA. .,Department of Neurology, University of Pennsylvania, Perelman School of Medicine, Philadelphia, PA, USA.
| | - Ralf A Husain
- Department of Neuropediatrics, Jena University Hospital, Jena, Germany.
| |
Collapse
|
49
|
Ni M, Black LF, Pan C, Vu H, Pei J, Ko B, Cai L, Solmonson A, Yang C, Nugent KM, Grishin NV, Xing C, Roeder E, DeBerardinis RJ. Metabolic impact of pathogenic variants in the mitochondrial glutamyl-tRNA synthetase EARS2. J Inherit Metab Dis 2021; 44:949-960. [PMID: 33855712 PMCID: PMC9219168 DOI: 10.1002/jimd.12387] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Revised: 04/06/2021] [Accepted: 04/12/2021] [Indexed: 12/15/2022]
Abstract
Glutamyl-tRNA synthetase 2 (encoded by EARS2) is a mitochondrial aminoacyl-tRNA synthetase required to translate the 13 subunits of the electron transport chain encoded by the mitochondrial DNA. Pathogenic EARS2 variants cause combined oxidative phosphorylation deficiency, subtype 12 (COXPD12), an autosomal recessive disorder involving lactic acidosis, intellectual disability, and other features of mitochondrial compromise. Patients with EARS2 deficiency present with variable phenotypes ranging from neonatal lethality to a mitigated disease with clinical improvement in early childhood. Here, we report a neonate homozygous for a rare pathogenic variant in EARS2 (c.949G>T; p.G317C). Metabolomics in primary fibroblasts from this patient revealed expected abnormalities in TCA cycle metabolites, as well as numerous changes in purine, pyrimidine, and fatty acid metabolism. To examine genotype-phenotype correlations in COXPD12, we compared the metabolic impact of reconstituting these fibroblasts with wild-type EARS2 versus four additional EARS2 variants from COXPD12 patients with varying clinical severity. Metabolomics identified a group of signature metabolites, mostly from the TCA cycle and amino acid metabolism, that discriminate between EARS2 variants causing relatively mild and severe COXPD12. Taken together, these findings indicate that metabolomics in patient-derived fibroblasts may help establish genotype-phenotype correlations in EARS2 deficiency and likely other mitochondrial disorders.
Collapse
Affiliation(s)
- Min Ni
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lauren F. Black
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chunxiao Pan
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hieu Vu
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Jimin Pei
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Bookyung Ko
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ling Cai
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
- Quantitative Biomedical Research Center, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Ashley Solmonson
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chendong Yang
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Nick V. Grishin
- Department of Biophysics, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Biochemistry, The University of Texas Southwestern Medical Center, Dallas, Texas
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
| | - Chao Xing
- Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Bioinformatics, The University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Ralph J. DeBerardinis
- Children’s Medical Center Research Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
- Department of Pediatrics, The University of Texas Southwestern Medical Center, Dallas, Texas
- Howard Hughes Medical Institute, The University of Texas Southwestern Medical Center, Dallas, Texas
- Eugene McDermott Center for Human Growth and Development, The University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
50
|
Treatment of ARS deficiencies with specific amino acids. Genet Med 2021; 23:2202-2207. [PMID: 34194004 PMCID: PMC8244667 DOI: 10.1038/s41436-021-01249-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Revised: 06/07/2021] [Accepted: 06/08/2021] [Indexed: 02/01/2023] Open
Abstract
Purpose Recessive cytosolic aminoacyl-tRNA synthetase (ARS) deficiencies are severe multiorgan diseases, with limited treatment options. By loading transfer RNAs (tRNAs) with their cognate amino acids, ARS are essential for protein translation. However, it remains unknown why ARS deficiencies lead to specific symptoms, especially early life and during infections. We set out to increase pathophysiological insight and improve therapeutic possibilities. Methods In fibroblasts from patients with isoleucyl-RS (IARS), leucyl-RS (LARS), phenylalanyl-RS-beta-subunit (FARSB), and seryl-RS (SARS) deficiencies, we investigated aminoacylation activity, thermostability, and sensitivity to ARS-specific amino acid concentrations, and developed personalized treatments. Results Aminoacylation activity was reduced in all patients, and further diminished at 38.5/40 °C (PLARS and PFARSB), consistent with infectious deteriorations. With lower cognate amino acid concentrations, patient fibroblast growth was severely affected. To prevent local and/or temporal deficiencies, we treated patients with corresponding amino acids (follow-up: 1/2–2 2/3rd years), and intensified treatment during infections. All patients showed beneficial treatment effects, most strikingly in growth (without tube feeding), head circumference, development, coping with infections, and oxygen dependency. Conclusion For these four ARS deficiencies, we observed a common disease mechanism of episodic insufficient aminoacylation to meet translational demands and illustrate the power of amino acid supplementation for the expanding ARS patient group. Moreover, we provide a strategy for personalized preclinical functional evaluation. ![]()
Collapse
|