1
|
Li S, Zhang C, Han R. An oversized AAV8 vector to deliver CPS1. MOLECULAR THERAPY. NUCLEIC ACIDS 2025; 36:102504. [PMID: 40206657 PMCID: PMC11979459 DOI: 10.1016/j.omtn.2025.102504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/11/2025]
Affiliation(s)
- Shuang Li
- Department of Pediatrics, Department of Medical and Molecular Genetics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Chen Zhang
- Department of Pediatrics, Department of Medical and Molecular Genetics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Renzhi Han
- Department of Pediatrics, Department of Medical and Molecular Genetics, Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| |
Collapse
|
2
|
Kohnz RA, Zhou D, Lou B, Yao H, McKenney D, Dokwal D, Villanueva R, Kocalis H, Ballard JE, Piesvaux J, Previs SF. Elucidation of Mechanism of Action in Drug Invention: Using Stable Isotope Tracers to Unravel Biochemical Kinetics. Pharmacol Res Perspect 2025; 13:e70099. [PMID: 40281645 PMCID: PMC12031654 DOI: 10.1002/prp2.70099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 03/25/2025] [Accepted: 03/27/2025] [Indexed: 04/29/2025] Open
Abstract
The invention of a therapeutic begins by characterizing features that differentiate healthy versus diseased states; this often presents as changes in the concentration of an analyte. Examples include elevated blood glucose in diabetes, high cholesterol in heart disease, and protein aggregation in neurodegeneration. Analyte concentrations reflect the (im)balance of synthetic and degradation rates; as such, aberrant biochemical kinetics drive the changes in endpoint concentration that define disease biology. Therapeutics aim to reset the concentration of a disease marker via modulation of biochemical kinetics. This is easy to understand for drugs directly targeting an enzyme in a pathway but, although less obvious, this can also be at the core of protein: protein interactions. For instance, stimulation of the insulin receptor changes the flux of several biochemical substrates (across multiple tissues); similarly, modulation of proprotein convertase subtilisin/kexin type 9-low density lipoprotein (PCSK9-LDL) receptor interactions alters cholesterol trafficking. These classic examples underscore the importance of studying biochemical kinetics at a clinical level. Here, we discuss how kinetic studies link disease biology with mechanism of action elucidation and screening. This has an immediate impact on (i) enabling in vitro-in vivo correlations in early discovery, (ii) enhancing exposure-response models aiding in human dose prediction, and (iii) providing support for biomarker plans, including clinical diagnostics. Mechanism of action studies can also influence modality selection; e.g., knowledge regarding target kinetics is needed when making decisions surrounding the development of a reversible inhibitor vs. an irreversible covalent modifier, or an intervention that affects target levels such as those which enhance protein degradation or reduce protein synthesis.
Collapse
Affiliation(s)
| | - Dan Zhou
- Merck & co., Inc.West PointPennsylvaniaUSA
| | - Bin Lou
- Merck & co., Inc.South San FranciscoCaliforniaUSA
| | | | | | | | | | | | | | | | | |
Collapse
|
3
|
Musunuru K, Grandinette SA, Wang X, Hudson TR, Briseno K, Berry AM, Hacker JL, Hsu A, Silverstein RA, Hille LT, Ogul AN, Robinson-Garvin NA, Small JC, McCague S, Burke SM, Wright CM, Bick S, Indurthi V, Sharma S, Jepperson M, Vakulskas CA, Collingwood M, Keogh K, Jacobi A, Sturgeon M, Brommel C, Schmaljohn E, Kurgan G, Osborne T, Zhang H, Kinney K, Rettig G, Barbosa CJ, Semple SC, Tam YK, Lutz C, George LA, Kleinstiver BP, Liu DR, Ng K, Kassim SH, Giannikopoulos P, Alameh MG, Urnov FD, Ahrens-Nicklas RC. Patient-Specific In Vivo Gene Editing to Treat a Rare Genetic Disease. N Engl J Med 2025. [PMID: 40373211 DOI: 10.1056/nejmoa2504747] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/17/2025]
Abstract
Base editors can correct disease-causing genetic variants. After a neonate had received a diagnosis of severe carbamoyl-phosphate synthetase 1 deficiency, a disease with an estimated 50% mortality in early infancy, we immediately began to develop a customized lipid nanoparticle-delivered base-editing therapy. After regulatory approval had been obtained for the therapy, the patient received two infusions at approximately 7 and 8 months of age. In the 7 weeks after the initial infusion, the patient was able to receive an increased amount of dietary protein and a reduced dose of a nitrogen-scavenger medication to half the starting dose, without unacceptable adverse events and despite viral illnesses. No serious adverse events occurred. Longer follow-up is warranted to assess safety and efficacy. (Funded by the National Institutes of Health and others.).
Collapse
Affiliation(s)
- Kiran Musunuru
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | | | - Xiao Wang
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Taylor R Hudson
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | - Kevin Briseno
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | - Anne Marie Berry
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Julia L Hacker
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Alvin Hsu
- Broad Institute of MIT and Harvard, Harvard University, Howard Hughes Medical Institute, Cambridge, MA
| | | | - Logan T Hille
- Massachusetts General Hospital-Harvard Medical School, Boston
| | - Aysel N Ogul
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | | | | | | | | | | | - Sarah Bick
- Children's Hospital of Philadelphia, Philadelphia
| | | | | | | | | | | | | | | | | | | | | | | | | | - He Zhang
- Integrated DNA Technologies, Coralville, IA
| | | | | | | | | | - Ying K Tam
- Acuitas Therapeutics, Vancouver, BC, Canada
| | | | - Lindsey A George
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | | | - David R Liu
- Broad Institute of MIT and Harvard, Harvard University, Howard Hughes Medical Institute, Cambridge, MA
| | - Kim Ng
- Children's Hospital of Philadelphia, Philadelphia
| | | | - Petros Giannikopoulos
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
- University of California, San Francisco, San Francisco
| | - Mohamad-Gabriel Alameh
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| | - Fyodor D Urnov
- Innovative Genomics Institute, University of California, Berkeley, Berkeley
| | - Rebecca C Ahrens-Nicklas
- Children's Hospital of Philadelphia, Philadelphia
- Perelman School of Medicine at the University of Pennsylvania, Philadelphia
| |
Collapse
|
4
|
Allegri G, Poms M, Zürcher N, Rüfenacht V, Rimann N, Mathis D, Thöny B, Gautschi M, Husain RA, Karall D, Orchel-Szastak K, Porta F, Roland D, Siri B, Dionisi-Vici C, Santer R, Häberle J. Characterization and treatment monitoring of ureagenesis disorders using stable isotopes. NPJ METABOLIC HEALTH AND DISEASE 2025; 3:19. [PMID: 40343092 PMCID: PMC12055570 DOI: 10.1038/s44324-025-00051-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 01/28/2025] [Indexed: 05/11/2025]
Abstract
Urea cycle disorders (UCDs) are a group of rare conditions, possibly life-threatening and without definitive cure besides liver transplantation. Traditional biochemical analyses/biomarkers cannot reliably determine changes in the UC-function from baseline to post-intervention. We describe a UHPLC-HRMS method to assess ureagenesis in plasma and dried blood spots for [15N]urea and [15N]amino acids, using [15N]ammonium chloride as tracer. [15N]enrichment of urea and amino acids was studied in controls (n = 22) and patients (n = 59), the latter showing characteristic ureagenesis variations according to their underlying metabolic defect. Follow-up of therapies was successful, as we observed restoration of [15N]urea production and lowering of [15N]glutamine. There were no adverse events, and only minimal amounts of tracer and samples required with a short sample preparation time and analysis. Thus, the method proved to be safe and efficient to monitor UCD patients of variable severity pre- and post-therapy, being suitable as physiological endpoint for development of therapies.
Collapse
Affiliation(s)
- Gabriella Allegri
- Division of Metabolism and Children’s Research Centre (CRC), University Children’s Hospital Zurich, Zurich, Switzerland
| | - Martin Poms
- Division Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Nadia Zürcher
- Division of Metabolism and Children’s Research Centre (CRC), University Children’s Hospital Zurich, Zurich, Switzerland
| | - Véronique Rüfenacht
- Division of Metabolism and Children’s Research Centre (CRC), University Children’s Hospital Zurich, Zurich, Switzerland
| | - Nicole Rimann
- Division of Metabolism and Children’s Research Centre (CRC), University Children’s Hospital Zurich, Zurich, Switzerland
| | - Déborah Mathis
- Division Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich, Switzerland
- University Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Beat Thöny
- Division of Metabolism and Children’s Research Centre (CRC), University Children’s Hospital Zurich, Zurich, Switzerland
- Division Clinical Chemistry and Biochemistry, University Children’s Hospital Zurich, Zurich, Switzerland
| | - Matthias Gautschi
- Division of Paediatric Endocrinology, Diabetology and Metabolism, Department of Paediatrics, and Institute of Clinical Chemistry, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Ralf A. Husain
- Centre for Inborn Metabolic Disorders, Department of Neuropediatrics, Jena University Hospital, Jena, Germany
| | - Daniela Karall
- Medical University of Innsbruck, Clinic for Paediatrics, Division of Inherited Metabolic Disorders, Innsbruck, Austria
| | - Karolina Orchel-Szastak
- Department of Paediatrics, Rheumatology and Rare Diseases, University Children’s Hospital, Krakow, Poland
| | - Francesco Porta
- Department of Pediatrics, AOU Citta della Salute e della Scienza, University of Torino, Turin, Italy
| | - Dominique Roland
- Centre des Maladies Héréditaires du Métabolisme, Département de Génétique Humaine, Institut de Pathologie et de Génétique, Gosselies, Belgium
| | - Barbara Siri
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - Carlo Dionisi-Vici
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children’s Hospital IRCCS, Rome, Italy
| | - René Santer
- Department of Pediatrics, University Medical Center Eppendorf, Hamburg, Germany
| | - Johannes Häberle
- Division of Metabolism and Children’s Research Centre (CRC), University Children’s Hospital Zurich, Zurich, Switzerland
| |
Collapse
|
5
|
Das AM. Urea cycle defects in adulthood: clinical presentation, diagnosis and treatment in genetically encoded hepatic metabolic disorders with a potential for encephalopathy. Metab Brain Dis 2025; 40:192. [PMID: 40285952 PMCID: PMC12033206 DOI: 10.1007/s11011-025-01619-5] [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: 03/16/2025] [Accepted: 04/19/2025] [Indexed: 04/29/2025]
Abstract
Hyperammonaemia is an important cause for encephalopathy. Ammonia is the waste product of amino acid degradation and cannot be excreted via urine. Ammonia is metabolized to water-soluble urea via the urea cycle. Hyperammonaemia not only occurs during acute liver failure, but also in rare genetically determined defects of enzymes or transporters involved in the urea cycle resulting in elevated ammonia concentrations. Enzyme defects include deficiency of carbamylphosphate synthase, N-acetylglutamate synthase, ornithine transcarbamylase, argininosuccinate lyase and arginase, transporter defects are citrin deficiency and HHH-syndrome. These urea cycle defects (UCD) mostly manifest for the first time during the neonatal period, infancy or childhood, however first clinical manifestations including encephalopathy may be observed in adulthood in milder forms. Therefore, physicians treating adults should be aware of clinical symptoms in UCD to make a timely diagnosis and initiate treatment. In adulthood, clinical symptoms are often uncharacteristic including headache, avoidance of high-protein food, psychiatric symptoms triggered by heavy exercise or delivery of a child, autism, attention deficit, lethargy, developmental delay and epilepsy. Elevated ammonia concentrations in blood are the biochemical hallmark. Some UCDs can be diagnosed at metabolite level, others only at genetic level. Treatment consists of eucaloric, low-protein diet supplemented with essential amino acids and vitamins/trace elements, and intake of arginine or citrulline. Pharmacological scavengers of nitrogen are benzoate and butyrate. If conservative therapy fails, hemodialysis should be considered. Prompt treatment during acute crises is essential for optimal outcome. Liver transplantation is considered in metabolically unstable patients. For arginase deficiency, enzyme replacement therapy is available.
Collapse
Affiliation(s)
- Anibh Martin Das
- Department of Paediatric Kidney, Liver and Metabolic Diseases, Hannover Medical School, Hannover, Germany.
- Hannover Medical School, Carl Neuberg Str. 1, D- 30625, Hannover, Germany.
| |
Collapse
|
6
|
Berardo C, Vasco A, Mauri A, Lucchi S, Cappelletti L, Saielli L, Rizzetto M, Biganzoli D, Montrasio C, Postorivo D, Pratiffi E, Meta A, Carelli S, Amorosi A, Paci S, Cefalo G, Furlan F, Menni F, Gasperini S, Crescitelli V, Banderali G, Zuccotti G, Alberti L, Cereda C. Expanded Newborn Screening in Italy: The First Report of Lombardy Region. Int J Neonatal Screen 2025; 11:31. [PMID: 40407514 PMCID: PMC12101260 DOI: 10.3390/ijns11020031] [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: 02/04/2025] [Revised: 03/28/2025] [Accepted: 04/23/2025] [Indexed: 05/26/2025] Open
Abstract
BACKGROUND Newborn screening (NBS) is a preventive healthcare program aiming at identifying the inborn errors of metabolism (IEMs) in asymptomatic infants to reduce the risk of severe complications. The aim of this study was to report the first years (2016-2020) of the expanded NBS program in the Lombardy region, Italy. METHODS Dried blood spots were collected from newborns' heels at 48-72 h after birth. FIA-MS/MS was performed to evaluate specific biochemical markers. Genetic confirmation was achieved via Sanger or NGS on newborns and reported to a clinical reference center (CRC). RESULTS A total of 343,507 newborns were tested; 1414/343,507 resulted as positive to NBS and were reported to the CRC. A total of 209 newborns were diagnosed with IEMs: 206 infants received a diagnosis of IEM through NBS, confirmed by genetic analysis; three neonates were not positive to NBS but were subsequentially diagnosed with IEMs. A total of 1208/343,507 were false positive cases. Twenty-seven types of IEMs were diagnosed in 209 patients: 111 newborns were affected by aminoacidemias, 11 by urea cycle disorders, 27 by organic acidemias, 34 by fatty acid oxidation disorders, and 26 by secondary conditions. CONCLUSIONS We report here for the first time the IEM incidence and distribution in the Lombardy region in the first five years of NBS.
Collapse
Affiliation(s)
- Clarissa Berardo
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy; (C.B.); (A.M.); (S.C.); (G.Z.)
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Alessandra Vasco
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Alessia Mauri
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy; (C.B.); (A.M.); (S.C.); (G.Z.)
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Simona Lucchi
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Laura Cappelletti
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Laura Saielli
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Manuela Rizzetto
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Davide Biganzoli
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Cristina Montrasio
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Diana Postorivo
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Elisa Pratiffi
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Andrea Meta
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Stephana Carelli
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy; (C.B.); (A.M.); (S.C.); (G.Z.)
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Alessandro Amorosi
- Welfare General Directorate, Regione Lombardia, Palazzo Lombardia, Piazza Città di Lombardia, 1, 20124 Milan, Italy;
| | - Sabrina Paci
- Clinical Department of Pediatrics, ASST Santi Paolo e Carlo Hospital, University of Milan, 20142 Milan, Italy; (S.P.); (G.C.); (G.B.)
| | - Graziella Cefalo
- Clinical Department of Pediatrics, ASST Santi Paolo e Carlo Hospital, University of Milan, 20142 Milan, Italy; (S.P.); (G.C.); (G.B.)
| | - Francesca Furlan
- Pediatric Intensive Care Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.F.); (F.M.)
| | - Francesca Menni
- Pediatric Intensive Care Unit, Fondazione IRCCS Cà Granda Ospedale Maggiore Policlinico, 20122 Milan, Italy; (F.F.); (F.M.)
| | - Serena Gasperini
- Pediatric Rare Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (S.G.); (V.C.)
| | - Viola Crescitelli
- Pediatric Rare Diseases Unit, Fondazione IRCCS San Gerardo dei Tintori, 20900 Monza, Italy; (S.G.); (V.C.)
| | - Giuseppe Banderali
- Clinical Department of Pediatrics, ASST Santi Paolo e Carlo Hospital, University of Milan, 20142 Milan, Italy; (S.P.); (G.C.); (G.B.)
| | - Gianvincenzo Zuccotti
- Pediatric Research Center “Romeo ed Enrica Invernizzi”, Department of Biomedical and Clinical Sciences, University of Milan, 20157 Milan, Italy; (C.B.); (A.M.); (S.C.); (G.Z.)
- Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy
| | - Luisella Alberti
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| | - Cristina Cereda
- Center of Functional Genomics and Rare diseases, Department of Pediatrics, Buzzi Children’s Hospital, 20154 Milan, Italy; (A.V.); (S.L.); (L.C.); (L.S.); (M.R.); (D.B.); (C.M.); (D.P.); (E.P.); (A.M.); (L.A.)
| |
Collapse
|
7
|
Burlina A, Ardissone A, Battini R, Burlina A, Gasperini S, Pession A, Porta F, Vici CD. Arginase 1 deficiency: a treatable form of spastic paraplegia. Neurol Sci 2025:10.1007/s10072-025-08153-3. [PMID: 40237972 DOI: 10.1007/s10072-025-08153-3] [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: 11/29/2024] [Accepted: 03/25/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Arginase 1 deficiency (ARG1-D) is a rare hereditary urea cycle disorder characterized by elevated arginine levels, resulting in progressive neurological impairment and severe physical and cognitive disability. Due to its low prevalence, overlapping symptoms with other neurological disorders, and current limitations in newborn screening tools, ARG1-D is often misdiagnosed or diagnosed late, limiting access to early interventions. AIM This review and expert opinion aim to provide an overview of the clinical manifestations, diagnostic challenges, and treatment options for ARG1-D, offering a practical resource for specialists to recognize this rare, progressive, yet treatable disease. RESULTS ARG1-D typically presents with progressive spastic paraplegia, developmental delays, cognitive impairment, and seizures, with symptom onset and severity varying by age. Differential diagnoses mainly include hereditary spastic paraplegia, cerebral palsy, and hyperornithinemia-hyperammonemia-homocitrullinuria syndrome, each with distinct clinical features and biochemical markers. Potential red flags for ARG1-D include elevated plasma arginine levels, spasticity, seizures, and cognitive impairment. These should prompt further examinations to confirm the diagnosis, which is based on biochemical assays for hyperargininemia and on genetic testing. Once confirmed, early treatment is advised, including dietary protein restriction, ammonia scavengers, antiepileptic drugs, and novel therapies, such as pegzilarginase, which targets the disease's metabolic root. CONCLUSION Experts stress the importance of increased awareness of ARG1-D characteristics, noting that early recognition and treatment are crucial to patient outcomes. Greater recognition of ARG1-D's distinctive features, differential diagnosis, and diagnostic tools, even among non-specialist clinicians, could help prevent misdiagnoses and facilitate the identification of this rare yet treatable condition.
Collapse
Affiliation(s)
- Alessandro Burlina
- Dept. of Medicine, Neurology Unit, St. Bassiano Hospital, Via dei Lotti 40, 36061, Bassano del Grappa, Italy.
| | - Anna Ardissone
- Child Neurology Unit, Department of Pediatric Neurosciences, Fondazione IRCCS Istituto Neurologico Carlo Besta, 20133, Milan, Italy
| | - Roberta Battini
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
- Department of Neuroscience, IRCCS Stella Maris Foundation, Calambrone, Pisa, Italy
| | - Alberto Burlina
- Division of Inherited Metabolic Diseases, Department of Woman's and Child's Health, University Hospital, 35129, Padua, Italy
| | - Serena Gasperini
- Department of Pediatrics, Fondazione IRCCS San Gerardo dei Tintori, Monza, Italy
| | - Andrea Pession
- Pediatric Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138, Bologna, Italy
| | - Francesco Porta
- Department of Pediatrics, AOU Citta' della Salute e della Scienza di Torino, 10126, Torino, Italy
| | - Carlo Dionisi Vici
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Childrens Hospital IRCCS, Rome, Italy
| |
Collapse
|
8
|
Ismet S, Hehsan MR. Perioperative Management of Argininemia in a Child Undergoing Circumcision: A Case Report. J Perianesth Nurs 2025; 40:251-253. [PMID: 39217526 DOI: 10.1016/j.jopan.2024.05.025] [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: 02/12/2024] [Revised: 05/21/2024] [Accepted: 05/22/2024] [Indexed: 09/04/2024]
Abstract
Argininemia is a rare autosomal recessive metabolic disorder characterized by a deficiency of arginase, a vital enzyme in the urea cycle. This metabolic defect results in the accumulation of arginine and its metabolites, leading to hyperammonemia and associated neurological symptoms. We present a case detailing the perioperative management of an 11-year-old male child diagnosed with argininemia undergoing circumcision. The perioperative management of patients with argininemia presents unique challenges due to the risk of hyperammonemia and neurological decompensation triggered by physiological stress, fasting, and the catabolic state associated with surgery. This case report highlights the importance of individualized anesthetic strategies for patients with rare metabolic disorders like argininemia. A multidisciplinary approach involving collaboration among anesthesiologists, endocrinologists, dietitians, and surgeons is essential to ensuring a safe perioperative experience for these patients. Further research is essential to refine perioperative protocols and optimal anesthetic interventions for individuals with argininemia undergoing surgical procedures.
Collapse
Affiliation(s)
- Suki Ismet
- Department of Anaesthesiology and Critical Care, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia
| | - Muhamad Rafiqi Hehsan
- Department of Anaesthesiology and Critical Care, School of Medical Sciences, Health Campus, Universiti Sains Malaysia, Kubang Kerian, Kelantan, Malaysia; Faculty of Medicine & Health Sciences, Universiti Sains Islam Malaysia, Nilai, Negeri Sembilan, Malaysia.
| |
Collapse
|
9
|
Martín-Hernández E, Bellusci M, Pérez-Mohand P, Correcher Medina P, Blasco-Alonso J, Morais-López A, de las Heras J, Meavilla Olivas SM, Dougherty-de Miguel L, Couce ML, Villarroya EC, García Jiménez MC, Moreno-Lozano PJ, Vives I, Gil-Campos M, Stanescu S, Ceberio-Hualde L, Camprodón M, Cortès-Saladelafont E, López-Urdiales R, Murray Hurtado M, Márquez Armenteros AM, Sierra Córcoles C, Peña-Quintana L, Ruiz-Pons M, Alcalde C, Castellanos-Pinedo F, Dios E, Barrio-Carreras D, Martín-Cazaña M, García-Peris M, Andrade JD, García-Volpe C, de los Santos M, García-Cazorla A, del Toro M, Felipe-Rucián A, Comino Monroy MJ, Sánchez-Pintos P, Matas A, Gil Ortega D, Martín-Rivada Á, Bergua A, Belanger-Quintana A, Vitoria I, Yahyaoui R, Pérez B, Morales-Conejo M, Quijada-Fraile P. Understanding the Natural History and the Effects of Current Therapeutic Strategies on Urea Cycle Disorders: Insights from the UCD Spanish Registry. Nutrients 2025; 17:1173. [PMID: 40218931 PMCID: PMC11990916 DOI: 10.3390/nu17071173] [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: 03/06/2025] [Revised: 03/25/2025] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
Background/Objectives: The present study updates the Spanish registry of patients with urea cycle disorders (UCD), originally established in 2013, to provide comprehensive epidemiological data and evaluate the impact of therapeutic strategies and newborn screening (NBS) on clinical outcomes. Methods: This retrospective, multicenter study focuses on 255 Spanish UCD patients. It includes all living and deceased cases up to February 2024, analyzing demographic, clinical, and biochemical variables. Results: The incidence of UCD in Spain over the past decade was 1:36,063 births. The most common defects were ornithine transcarbamylase deficiency (OTCD) and argininosuccinate synthetase deficiency. Early-onset (EO) cases comprised 32.7%, and 10.6% were diagnosed through NBS. Global mortality was 14.9%, higher in carbamoylphosphate synthetase 1 deficiency (36.8%) and male OTCD patients (32.1%) compared to other defects (p = 0.013). EO cases presented a higher mortality rate (35.8%) than late-onset (LO) cases (7.1%) (p < 0.0001). The median ammonia level in deceased patients was higher at 1058 µmol/L (IQR 410-1793) than in survivors at 294 µmol/L (IQR 71-494) (p < 0.0001). Diagnosis through NBS improved survival and reduced neurological impairment compared to symptomatic diagnosis. Neurological impairment occurred in 44% of patients, with worse neurological outcomes observed in patients with argininosuccinate lyase deficiency, arginase 1 deficiency, hyperornithinemia-hyperammonemia-homocitrullinuria, EO presentations, pre-2014 diagnosis, and patients with higher levels of ammonia at diagnosis. Among transplanted patients (20.6%), survival was 95.2%, with no significant neurological differences compared to non-transplanted patients. Conclusions: This updated analysis highlights the positive impact of NBS and advanced treatments on mortality and neurologic outcomes. Persistent neurological challenges underscore the need for further therapeutic strategies.
Collapse
Affiliation(s)
- Elena Martín-Hernández
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Marcello Bellusci
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Patricia Pérez-Mohand
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Patricia Correcher Medina
- Unidad de Nutrición y Metabolopatías, Hospital Universitario La Fé, 46026 Valencia, Spain; (P.C.M.); (M.G.-P.); (I.V.)
| | - Javier Blasco-Alonso
- Unidad de Gastroenterología, Hepatología y Nutrición Pediátrica, Laboratorio de Bioquímica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.B.-A.); (R.Y.)
| | - Ana Morais-López
- Unidad de Nutrición Infantil y Enfermedades Metabólicas, Hospital Universitario La Paz, 28046 Madrid, Spain; (A.M.-L.); (J.D.A.); (A.B.)
| | - Javier de las Heras
- Hereditary Metabolic Diseases Unit, Hospital Universitario Cruces, MetabERN, Biobizkaia Health Research Institute, 48903 Bilbao, Spain; (J.d.l.H.); (L.C.-H.)
| | - Silvia María Meavilla Olivas
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Lucy Dougherty-de Miguel
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - Maria Luz Couce
- Unidad de Enfermedades Metabólicas, Hospital Clínico Universitario de Santiago, MetabERN, IDIS, 15706 Santiago de Compostela, Spain; (M.L.C.); (P.S.-P.)
| | - Elvira Cañedo Villarroya
- Unidad de Nutrición y Enfermedades Metabólicas, Hospital Universitario Niño Jesús, 28009 Madrid, Spain;
| | | | - Pedro Juan Moreno-Lozano
- Unidad de Errores Congénitos del Metabolismo en el Adulto, Medicina Interna, Hospital Clínic, 08036 Barcelona, Spain; (P.J.M.-L.); (A.M.)
| | - Inmaculada Vives
- Gastroenterología Pediátrica, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain; (I.V.); (D.G.O.)
| | - Mercedes Gil-Campos
- Unidad de Metabolismo, Hospital Universitario Reina Sofía, IMIBIC, UCO, 14004 Córdoba, Spain; (M.G.-C.); (M.J.C.M.)
| | - Sinziana Stanescu
- Unidad de Enfermedades Metabólicas, Hospital Ramón y Cajal, MetabERN, 28034 Madrid, Spain; (S.S.); (A.B.-Q.)
| | - Leticia Ceberio-Hualde
- Hereditary Metabolic Diseases Unit, Hospital Universitario Cruces, MetabERN, Biobizkaia Health Research Institute, 48903 Bilbao, Spain; (J.d.l.H.); (L.C.-H.)
| | - María Camprodón
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - Elisenda Cortès-Saladelafont
- Unidad de Neurología Pediátrica y Enfermedades Metabólicas, Hospital Germans Trias i Pujol, 08916 Barcelona, Spain;
| | - Rafael López-Urdiales
- Departamento de Endocrinología y Nutrición, Hospital de Bellvitge, 08907 Barcelona, Spain;
| | - Mercedes Murray Hurtado
- Unidad de Nutrición y Metabolopatías, Pediatría, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain; (M.M.H.); (Á.M.-R.)
| | | | | | - Luis Peña-Quintana
- Gastroenterología y Nutrición Pediátrica, Complejo Hospitalario Universitario Insular-Materno Infantil, CIBEROBN-ISCIII, Universidad de Las Palmas de Gran Canaria, 35016 Las Palmas de Gran Canaria, Spain;
| | - Mónica Ruiz-Pons
- Unidad de Nutrición y Enfermedades Metabólicas, Pediatría, Hospital Universitario Virgen de la Candelaria, 38010 Tenerife, Spain;
| | - Carlos Alcalde
- Servicio de Pediatría, Hospital Universitario Río Ortega, 47012 Valladolid, Spain;
| | | | - Elena Dios
- Endocrinología y Enfermedades Metabólicas, MetabERN, Hospital Virgen del Rocío, 41013 Sevilla, Spain;
| | - Delia Barrio-Carreras
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - María Martín-Cazaña
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Mónica García-Peris
- Unidad de Nutrición y Metabolopatías, Hospital Universitario La Fé, 46026 Valencia, Spain; (P.C.M.); (M.G.-P.); (I.V.)
| | - José David Andrade
- Unidad de Nutrición Infantil y Enfermedades Metabólicas, Hospital Universitario La Paz, 28046 Madrid, Spain; (A.M.-L.); (J.D.A.); (A.B.)
| | - Camila García-Volpe
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Mariela de los Santos
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Angels García-Cazorla
- Unidad de Enfermedades Metabólicas, Hospital San Joan de Déu, MetabERN, CIBERER, 08035 Barcelona, Spain; (S.M.M.O.); (C.G.-V.); (M.d.l.S.); (A.G.-C.)
| | - Mireia del Toro
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - Ana Felipe-Rucián
- Unidad de Enfermedades Metabólicas, Hospital Vall d’Hebrón, MetabERN, VHIR, 08035 Barcelona, Spain; (L.D.-d.M.); (M.C.); (M.d.T.); (A.F.-R.)
| | - María José Comino Monroy
- Unidad de Metabolismo, Hospital Universitario Reina Sofía, IMIBIC, UCO, 14004 Córdoba, Spain; (M.G.-C.); (M.J.C.M.)
| | - Paula Sánchez-Pintos
- Unidad de Enfermedades Metabólicas, Hospital Clínico Universitario de Santiago, MetabERN, IDIS, 15706 Santiago de Compostela, Spain; (M.L.C.); (P.S.-P.)
| | - Ana Matas
- Unidad de Errores Congénitos del Metabolismo en el Adulto, Medicina Interna, Hospital Clínic, 08036 Barcelona, Spain; (P.J.M.-L.); (A.M.)
| | - David Gil Ortega
- Gastroenterología Pediátrica, Hospital Clínico Universitario Virgen de la Arrixaca, 30120 Murcia, Spain; (I.V.); (D.G.O.)
| | - Álvaro Martín-Rivada
- Unidad de Nutrición y Metabolopatías, Pediatría, Hospital Universitario de Canarias, 38320 Santa Cruz de Tenerife, Spain; (M.M.H.); (Á.M.-R.)
| | - Ana Bergua
- Unidad de Nutrición Infantil y Enfermedades Metabólicas, Hospital Universitario La Paz, 28046 Madrid, Spain; (A.M.-L.); (J.D.A.); (A.B.)
| | - Amaya Belanger-Quintana
- Unidad de Enfermedades Metabólicas, Hospital Ramón y Cajal, MetabERN, 28034 Madrid, Spain; (S.S.); (A.B.-Q.)
| | - Isidro Vitoria
- Unidad de Nutrición y Metabolopatías, Hospital Universitario La Fé, 46026 Valencia, Spain; (P.C.M.); (M.G.-P.); (I.V.)
| | - Raquel Yahyaoui
- Unidad de Gastroenterología, Hepatología y Nutrición Pediátrica, Laboratorio de Bioquímica, Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain; (J.B.-A.); (R.Y.)
| | - Belén Pérez
- Centro de Diagnóstico de Enfermedades Moleculares, IdiPAZ, CIBERER, Universidad Autónoma Madrid, 28049 Madrid, Spain;
| | - Montserrat Morales-Conejo
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| | - Pilar Quijada-Fraile
- Unidad de Enfermedades Mitocondriales-Metabólicas Hereditarias, Hospital Universitario 12 de Octubre, Instituto de Investigación Imas12, MetabERN, CIBERER, 28041 Madrid, Spain; (M.B.); (P.P.-M.); (D.B.-C.); (M.M.-C.); (M.M.-C.); (P.Q.-F.)
| |
Collapse
|
10
|
Stepien KM, McSweeney M, Ochoa-Ferraro A, Vara R, Riley P, Smith M. Perspectives on long-term medical management of urea cycle disorders: insights from a survey of UK healthcare professionals. Orphanet J Rare Dis 2025; 20:135. [PMID: 40102865 PMCID: PMC11921535 DOI: 10.1186/s13023-025-03647-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 03/01/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Urea cycle disorders (UCDs) are rare inborn errors of metabolism which impact the body's ability to detoxify ammonia produced during protein metabolism. In the UK, there is a nationally adopted guideline for the emergency management of hyperammonaemia in UCD patients, however there is no guideline for long‑term management, and treatment decisions are left to the discretion of individual healthcare professionals (HCPs). RESULTS Twenty-three HCPs, comprising 13 (57%) metabolic consultants, two (9%) specialist nurses, four (17%) pharmacists, and four (17%) dietitians, participated in interviews to document their attitudes and beliefs regarding the long‑term management of UCD patients, including their current practices, treatment goals, and clinical ambitions. The highest priority for 14/23 (61%) of HCPs was to minimise the risk of hyperammonaemia, however the ammonia level that HCPs advised they aimed for varied significantly, with some targeting above the upper limit of normal. Glycerol phenylbutyrate was the highest ranked ammonia scavenger treatment amongst HCPs for safety, tolerability, duration of scavenging action and reducing patient burden, and HCPs suggested that it would be the first-line treatment in an updated guideline. All prescribing HCPs agreed they would prefer their patients receive a licenced product rather than an unlicensed one for reasons including more reliable supply, greater insurance/legitimacy, and the reassurance of regulatory scrutiny and approval. However, analysis of NHS England's dispensing data between July 2023 and June 2024 indicated annual spend on nitrogen scavengers of £6.7 million with unlicensed specials accounting for £3 million (45%) of the total. Differences between HCPs in the awareness of clinically relevant characteristics of ammonia scavengers, including their sodium and propylene glycol content, were observed. CONCLUSIONS To standardise the treatment of UCDs within and between metabolic centres in the UK, there is merit in developing a UK-specific treatment guideline.
Collapse
Affiliation(s)
- Karolina M Stepien
- Adult Inherited Metabolic Disease Department, Salford Care Organisation, Northern Care Alliance NHS Foundation Trust, Salford, M6 8HD, UK
| | - Melanie McSweeney
- Department of Paediatric Inherited Metabolic Disease, Great Ormond Street Hospital NHS Foundation Trust, London, WC1N 3JH, UK
| | - Antonio Ochoa-Ferraro
- Adult Inherited Metabolic Disease Service, University Hospitals Birmingham NHS Foundation Trust, Queen Elizabeth Hospital Birmingham, Edgbaston, Birmingham, B15 2GW, UK
| | - Roshni Vara
- Department of Paediatric Inherited Metabolic Disease, Evelina London Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, London, SE1 7EH, UK
| | - Paul Riley
- Nexcea, Glasshouse, Alderley Park, Macclesfield, SK10 4ZE, UK.
| | - Megan Smith
- Nexcea, Glasshouse, Alderley Park, Macclesfield, SK10 4ZE, UK
| |
Collapse
|
11
|
Zhang Y, Gu X, Shi C, Xiong H, Xiao D, Deng Z, Wang L, Yang X, Wei T, Liang P, Hao H. Clinical and biochemical characteristics of patients with ornithine transcarbamylase deficiency and in silico analysis of OTC gene. Orphanet J Rare Dis 2025; 20:131. [PMID: 40102887 PMCID: PMC11916849 DOI: 10.1186/s13023-025-03624-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 02/18/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND This study seeks to elucidate the clinical and biochemical features of Ornithine transcarbamylase deficiency (OTCD), a pleomorphic congenital hyperammonemia disorder with a non-specific clinical phenotype. Additionally, the research aims to analyze the mutation spectrum of the OTC gene and its potential association with phenotype, as well as to perform an in silico analysis of novel OTC variants to elucidate their structure-function relationship. METHODS In this study, we conducted a retrospective analysis of the clinical and biochemical features of 12 patients with OTCD and examined their metabolite profiles. Additionally, we reviewed existing literature to explore the range of mutations in the OTC gene and their possible associations with phenotypic outcomes. Furthermore, we employed the high ambiguity-driven protein-protein docking (HADDOCK) algorithm and protein-ligand interaction profiler (PLIP) to predict the pathogenicity of these mutations and elucidate the underlying mechanisms of pathogenesis in novel variants of the OTC gene. RESULTS Nine cases, all of which were male, presented with early onset, while two cases, all of which were female, exhibited late onset. Additionally, one male case was asymptomatic. The ages of the patients at the time of diagnosis ranged from 1 day to 12 years. Peak plasma ammonia levels were found to be higher in patients with early onset compared to those with late onset. Molecular analyses identified a total of 12 different mutations, including two novel mutations (V323G and R320P). In silico analysis indicated a potential difference in affinity between wild-type and mutant OTCase, with V323G and R320P mutations leading to a decreased binding ability of OTCase to the substrate, potentially disrupting its function. CONCLUSION This study broadened the genetic variation spectrum of OTCD and provided substantial evidence for genetic counselling to affected families. Additionally, we elucidated variant data of OTC in Chinese patients through comprehensive literature review. Given the ongoing uncertainty surrounding the genotype-phenotype correlation of OTCD, the results of our in silico analysis can contribute to a deeper understanding of this complex, rare, and severe genetic disorder.
Collapse
Affiliation(s)
- YinChun Zhang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated HospitalSun Yat-sen University, Guangzhou, China
| | - Xia Gu
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Inborn Errors of Metabolism Laboratory, The Sixth Affiliated Hospital, Sun Yat sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated HospitalSun Yat-sen University, Guangzhou, China
| | - Congcong Shi
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Inborn Errors of Metabolism Laboratory, The Sixth Affiliated Hospital, Sun Yat sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated HospitalSun Yat-sen University, Guangzhou, China
| | - Hui Xiong
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated HospitalSun Yat-sen University, Guangzhou, China
| | - DongFan Xiao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated HospitalSun Yat-sen University, Guangzhou, China
| | - ZhiRong Deng
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lu Wang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - XiMei Yang
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tao Wei
- Department of Bioengineering, College of Food Science, South China Agricultural University, Guangzhou, Guangdong, 510642, China.
| | - PuPing Liang
- MOE Key Laboratory of Gene Function and Regulation, State Key Laboratory of Biocontrol, School of Life SciencesSun Yat-sen University, Guangzhou, Guangdong, 510275, China.
| | - Hu Hao
- Department of Pediatrics, The Sixth Affiliated Hospital, Sun Yat-sen University, No. 26 Yuancun Erheng Road, Tianhe District, Guangzhou, 510655, China.
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
- Inborn Errors of Metabolism Laboratory, The Sixth Affiliated Hospital, Sun Yat sen University, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated HospitalSun Yat-sen University, Guangzhou, China.
| |
Collapse
|
12
|
Zhou J, Liang S, Yin L, Frassetto A, Graham AR, White R, Principe M, Severson M, Palmer T, Naidu S, Jacquinet E, Zimmer M, Finn PF, Martini PGV. Characterization of a novel conditional knockout mouse model to assess efficacy of mRNA therapy in the context of severe OTC deficiency. Mol Ther 2025; 33:1197-1212. [PMID: 39799396 PMCID: PMC11897776 DOI: 10.1016/j.ymthe.2025.01.010] [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: 06/02/2024] [Revised: 11/21/2024] [Accepted: 01/08/2025] [Indexed: 01/15/2025] Open
Abstract
Ornithine transcarbamylase deficiency (OTCD) is the most common urea-cycle disorder, characterized by hyperammonemia and accompanied by a high unmet patient need. mRNA therapies have been shown to be efficacious in hypomorphic Sparse-fur abnormal skin and hair (Spf-ash) mice, a model of late-onset disease. However, studying the efficacy of ornithine transcarbamylase (OTC) mRNA therapy in traditional knockout mice, a model for severe early-onset OTCD, is hampered by the rapid lethality of the model and poor lipid nanoparticle (LNP) uptake into neonatal mouse liver. We developed a novel tamoxifen-inducible mouse to study the effect of mRNA therapy in the context of complete or near-complete OTC loss in adult animals. Characterization of the model showed that it is highly reproducible, 100% penetrant, and phenocopies hallmarks of human disease, with animals exhibiting decreased body weight, hyperammonemia, and brain edema. Delivery of OTC mRNA increased survival, maintained body weight, delayed the onset of hyperammonemia, and reduced brain edema. Therefore, this model provides a platform to study LNP-mediated mRNA therapies for the treatment of late-onset OTCD.
Collapse
Affiliation(s)
| | - Shi Liang
- Moderna, Inc., Cambridge, MA 02142, USA
| | - Ling Yin
- Moderna, Inc., Cambridge, MA 02142, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
13
|
Gemici Karaaslan B, Kiykim A, Burtecene N, Gokden M, Cansever MS, Hopurcuoglu D, Cengiz GN, Topcu B, Zubarioğlu T, Kiykim E, Cokuğras H, Aktuglu Zeybek AC. Amino Acid Metabolism and Immune Dysfunction in Urea Cycle Disorders: T and B Cell Perspectives. J Inherit Metab Dis 2025; 48:e70009. [PMID: 39957310 PMCID: PMC11831096 DOI: 10.1002/jimd.70009] [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: 10/07/2024] [Revised: 12/17/2024] [Accepted: 02/05/2025] [Indexed: 02/18/2025]
Abstract
Urea cycle disorders (UCDs) are a group of genetic metabolic conditions characterized by enzyme deficiencies responsible for detoxifying ammonia. Hyperammonemia, the accumulation of intermediate metabolites, and a deficiency of essential amino acids-due to a protein-restrictive diet and the use of ammonia scavengers-can increase the risk of infections, particularly during metabolic crises. While the underlying mechanisms of immune suppression are still being fully elucidated, hyperammonemia may impair the function of immune cells, particularly T cells and macrophages, inhibiting the proliferation of T cells and cytokine production. Arginine, which is essential for T-cell activation and function, may also be limited in these patients, and its depletion can increase their vulnerability to infections. Twenty-four UCD patients and 31 healthy donors were recruited for the study. Peripheral lymphocyte subset analysis, intracellular protein and cytokine staining, and proliferation assays were performed by flow cytometry. Amino acid levels were measured using the HPLC method. The UCD patients exhibited low lymphocyte-proliferation capacity in both proximal and distal defects in response to phytohaemagglutinin (PHA) and anti-CD2, anti-CD3, and anti-CD28 (CD-mix), which was lower than healthy controls. Proximal-UCD patients exhibited a significantly higher response for IFN-γ compared to both distal-UCD patients and healthy controls. The different amino acids in the culture medium were changed significantly in the groups. This study highlights significant immune dysfunctions in UCD patients, particularly impaired T-cell proliferation and altered amino acid metabolism. Proximal UCD patients exhibited a higher IFN-γ response, indicating a potential for hyperinflammation. Despite this, infection rates did not significantly differ between proximal UCD and distal UCD patients, although distal UCD patients had higher hospitalization rates. Amino acid analysis revealed distinct metabolic disruptions, emphasizing the complex interplay between metabolism and immune function. These findings suggest that UCDs cause profound immune alterations, necessitating further research to develop targeted therapeutic strategies.
Collapse
Affiliation(s)
- Betul Gemici Karaaslan
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Ayca Kiykim
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Nihan Burtecene
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Meltem Gokden
- Cerrahpasa Medical FacultyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Mehmet Serif Cansever
- Department of Medical Services and TechniquesVocational School of Health Services, Istanbul University‐CerrahpaşaIstanbulTürkiye
| | - Duhan Hopurcuoglu
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Nutrition and MetabolismIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Gökçe Nuran Cengiz
- Cerrahpasa Medical Faculty, Department of PediatricsIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Birol Topcu
- Department of BiostatisticNamik Kemal UniversityTekirdagTürkiye
| | - Tanyel Zubarioğlu
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Nutrition and MetabolismIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Ertugrul Kiykim
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Nutrition and MetabolismIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Haluk Cokuğras
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Immunology and AllergyIstanbul University‐CerrahpasaIstanbulTürkiye
| | - Ayse Cigdem Aktuglu Zeybek
- Cerrahpasa Medical Faculty, Department of Pediatrics, Division of Nutrition and MetabolismIstanbul University‐CerrahpasaIstanbulTürkiye
| |
Collapse
|
14
|
Li Y, Tian R, Wang D, Zhang H, Zhou Y, Ma C, Zhang H, Zhang K, Liu S. Potential role of ARG1 c.57G > A variant in Argininemia. Genes Genomics 2025; 47:197-205. [PMID: 39567422 DOI: 10.1007/s13258-024-01595-7] [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: 06/20/2024] [Accepted: 10/25/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND AND OBJECTIVE Argininemia (OMIM: 207800), as well as arginase deficiency, a disorder of the urea cycle caused by deficiency of arginase 1 (ARG1, NP_000036.2), is a scarce autosomal recessive genetic disease. The patients who suffered with argininemia often showed spastic paraplegia, epileptic seizures, severe mental retardation, and even the hyperammonemia. In neonatal screening, we found a healthy baby with mild elevated arginine levels. We have demonstrated the genetic etiology of the patient. METHODS The patient's clinical characteristic and family history were collected. The technologies including Next Generation Sequencing (NGS), Sanger sequencing, Bioinformatics Analysis, RNA extraction, cDNA obtained, Sanger sequencing, Minigene splicing assay, Real-time PCR, Single-molecule real-time (SMRT) sequencing were applied. RESULTS One homozygous variant, c.57G > A (p.Q19=), was identified in the proband, which was inherited from the parents. Through different detection methods, we found that the c.57G > A variant causes three different transcriptional versions: normal mRNA (mRNA from blood), mRNA with the exon2 deletion (73bp, mRNA from blood and minigene assay), and mRNA sequence from the SMRT sequencing (parts of exons and introns were detected, including exon 1-4, intron 1 and 4, and part of intron 2, 3, and 5). The expression of ARG1 mRNA and protein also decreased in the blood. The related genes of NMD (Nonsense-mediated mRNA decay), SMG1, UPF1, and UPF3b, were expressed higher than the controls in the blood, which hints the NMD could play a role in the mRNA decay regarding the cDNA with 73bp deletion by c.57G > A variant. CONCLUSIONS The study is the first study considering a synonymous variant of the ARG1 gene influencing alternative splicing(AS). Otherwise, the variant c.57G > A is relatively frequent in the general population( MAF = 0.0146). Our discovery revealed the variant possesses partial pathogenic potential, which would contribute to the deeper understanding and gold model for the intricate relationship between genetic mutations, arginine metabolism, and physical function.
Collapse
Affiliation(s)
- Yixiao Li
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, 250022, Shandong, China
- Department of Ophthalmology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250001, Shandong, China
- Department of Ophthalmology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250012, Shandong, China
| | - Rujin Tian
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, 250022, Shandong, China
| | - Dong Wang
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, 250022, Shandong, China
| | - Haozheng Zhang
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, 250022, Shandong, China
| | - Yi Zhou
- Department of Ophthalmology, Affiliated Hospital of Xuzhou Medical University, Xuzhou, 221002, Jiangsu, China
| | - Chunli Ma
- Shandong Academy of Medical Science, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Han Zhang
- Department of Ophthalmology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, 250001, Shandong, China.
| | - Kaihui Zhang
- Pediatric Research Institute, Children's Hospital Affiliated to Shandong University, Jinan, 250022, Shandong, China.
| | - Shu Liu
- Pediatric Endocrinology and Inherited Metabolic Department, Guangdong Women and Children Hospital, Guangzhou, 511442, Guangdong, China.
| |
Collapse
|
15
|
Kaya B, Akduman H, Dilli D, Geyik C, Karaman A, Uçan B, Kaya Ö, Aydoğan S, Zenciroğlu A. Rare Diseases and Syndromes Observed in Newborn Babies with Idiopathic Hypertrophic Pyloric Stenosis. Z Geburtshilfe Neonatol 2025; 229:53-59. [PMID: 39384293 DOI: 10.1055/a-2410-5830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/11/2024]
Abstract
AIM Although infantile hypertrophic pyloric stenosis is a multifactorial disease caused by genetic and environmental factors, the role of genetic factors has become more important recently. With this study, we aimed to present rare diseases accompanying infantile hypertrophic pyloric stenosis caused by genetic factors. PATIENTS AND METHODS This is a retrospective study. Babies who were operated on with the diagnosis of infantile hypertrophic pyloric stenosis in the neonatal intensive care unit between 2000 and 2022 and had additional diseases were included in the study. RESULTS 9.8% of patients diagnosed with infantile hypertrophic pyloric stenosis had an accompanying rare disease. CONCLUSION Early diagnosis and treatment of rare diseases associated with infantile hypertrophic pyloric stenosis are of great importance in reducing morbidity/mortality due to these diseases.
Collapse
Affiliation(s)
- Başak Kaya
- Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Hasan Akduman
- Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Dilek Dilli
- Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Cem Geyik
- Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Ayşe Karaman
- Pediatric Surgery, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Berna Uçan
- Pediatric Radiology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Özkan Kaya
- Pediatric Cardiology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Seda Aydoğan
- Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| | - Ayşegül Zenciroğlu
- Neonatology, SBU Ankara Dr. Sami Ulus Maternity Child Health and Diseases Training and Research Hospital, Ankara, Turkey
| |
Collapse
|
16
|
Stolwijk NN, Häberle J, Huidekoper HH, Wagenmakers MAEM, Hollak CEM, Bosch AM. Mapping challenges in the accessibility of treatment products for urea cycle disorders: A survey of European healthcare professionals. J Inherit Metab Dis 2025; 48:e12815. [PMID: 39625308 PMCID: PMC11670152 DOI: 10.1002/jimd.12815] [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: 08/30/2024] [Revised: 10/17/2024] [Accepted: 11/04/2024] [Indexed: 12/28/2024]
Abstract
Current management guidelines for urea cycle disorders (UCDs) offer clear strategies, incorporating both authorized and non-authorized medicinal products (including intravenous formulations and products regulated as food). These varying product categories are subject to specific accessibility challenges related to availability, reimbursement, and pricing. The aim of this study is to identify potential obstacles to optimal UCD treatment implementation in European clinical practice. A survey aimed at metabolic healthcare professionals (HCPs) managing patients with UCDs in Europe was disseminated through the European Reference Network for Hereditary Metabolic Disorders and the European registry and network for intoxication type metabolic diseases. Forty-eight survey responses were collected from 21 European countries. In 16 of these countries, at least one metabolic HCP reported challenges in accessing UCD products. Reimbursement issues were reported for most products (8/10), including both authorized and non-authorized products. Availability-related challenges were also reported for 8/10 products, although unavailability was limited to non-authorized products. Prices impacted accessibility for all authorized products (3/3) and one non-authorized IV product. The accessibility of UCD treatment products varied across Europe, although no clear regional variations could be discerned. Survey data revealed that metabolic HCPs experience challenges in accessing both authorized and non-authorized products for UCD management in the majority of European countries. This indicates that registering unauthorized products may not resolve all issues. Improved reimbursement policies and fair pricing models, as well as (adjusted) authorization procedures may help address these concerns, thereby optimizing treatment access for UCD patients.
Collapse
Affiliation(s)
- Nina N. Stolwijk
- Medicines for Society (Medicijn voor de Maatschappij)Platform at Amsterdam UMC ‐ University of AmsterdamAmsterdamThe Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for inborn errors of Metabolism, MetabERNUniversity of AmsterdamAmsterdamThe Netherlands
| | - Johannes Häberle
- Division of Metabolism and Children's Research CenterUniversity Children's Hospital ZurichZurichSwitzerland
| | - Hidde H. Huidekoper
- Department of Pediatrics, Center for Lysosomal and Metabolic Diseases, Erasmus MCUniversity Medical Center RotterdamRotterdamThe Netherlands
| | - Margreet A. E. M. Wagenmakers
- Department of Internal Medicine, Center for Lysosomal and Metabolic Diseases, Erasmus MCErasmus University Medical Centre RotterdamRotterdamNetherlands
| | - Carla E. M. Hollak
- Medicines for Society (Medicijn voor de Maatschappij)Platform at Amsterdam UMC ‐ University of AmsterdamAmsterdamThe Netherlands
- Department of Endocrinology and Metabolism, Amsterdam UMC, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM) Research Institute, Expertise Center for inborn errors of Metabolism, MetabERNUniversity of AmsterdamAmsterdamThe Netherlands
| | - Annet M. Bosch
- Department of Pediatrics, Division of Metabolic Diseases, Emma Children's Hospital, Amsterdam Gastroenterology Endocrinology Metabolism (AGEM)Amsterdam UMC location University of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
17
|
Gu X, Mo W, Zhuang G, Shi C, Wei T, Zhang J, Tu C, Cai Y, Liao B, Hao H. Visualization of argininosuccinate synthetase by in silico analysis: novel insights into citrullinemia type I disorders. Front Mol Biosci 2024; 11:1482773. [PMID: 39649700 PMCID: PMC11621003 DOI: 10.3389/fmolb.2024.1482773] [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: 08/20/2024] [Accepted: 10/21/2024] [Indexed: 12/11/2024] Open
Abstract
Background Citrullinemia type I disorders (CTLN1) is a genetic metabolic disease caused by argininosuccinate synthetase (ASS1) gene mutation. To date, the human genome mutation database has documented over 100 variants of the ASS1 gene. This study reported a novel deletion-insertion variant of ASS1 gene and employed various prediction tools to determine its pathogenicity. Methods We reported a case of early-onset CTLN1. Whole exome sequencing was conducted to identify genetic mutations. We employed various structure prediction tools to generate accurate 3D models and utilized computational biology tools to elucidate the disparities between the wild-type and mutant proteins. Results The patient was characterized by severe clinical manifestations, including poor responsiveness, lethargy, convulsions, and cardiac arrest. Notably, the patient exhibited significantly elevated blood ammonia levels (655 μmol/L; normal reference: 10-30 μmol/L) and increased citrulline concentrations (936 μmol/L; normal reference: 5-25 μmol/L). Whole exome sequencing revealed a in-frame deletion-insertion mutation c.1128_1134delinsG in the ASS1 gene of unknown significance, which has not been previously reported. Our finding indicated that the C- terminal helix domain of the mutant protein structure, which was an important structure for ASS1 protein to form protein tetramers, was indeed more unstable than that of the wild-type protein structure. Conclusion Through conducting an in silico analysis on this unique in-frame deletion-insertion variant of ASS1, our aim was to enhance understanding regarding its structure-function relationship as well as unraveling the molecular mechanism underlying CTLN1.
Collapse
Affiliation(s)
- Xia Gu
- Department of Neonatology, The Sixth Affiliated Hospital, Sun-Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenhui Mo
- Department of Neonatology, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Guiying Zhuang
- Department of Neonatology, The Maternal and Child Health Care Hospital of Huadu, Guangzhou, China
| | - Congcong Shi
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
- Inborn Errors of Metabolism Laboratory, The Sixth Affiliated Hospital, Sun-Yat-Sen University, Guangzhou, China
| | - Tao Wei
- Guangdong Shaohe Biotechnology Co., LTD., Guangzhou, China
| | - Jinze Zhang
- Guangdong Shaohe Biotechnology Co., LTD., Guangzhou, China
| | - Chiaowen Tu
- Department of Neonatology, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Yao Cai
- Department of Neonatology, The Sixth Affiliated Hospital, Sun-Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Biwen Liao
- Department of Neonatology, Foshan Fosun Chancheng Hospital, Foshan, China
| | - Hu Hao
- Department of Neonatology, The Sixth Affiliated Hospital, Sun-Yat-Sen University, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
18
|
Olofsson S, Löfvendahl S, Widén J, Jacobson L, Lindgren P, Stepien KM, Arnoux J, Luz Couce Pico M, Leão Teles E, Rudebeck M. Disease burden among patients with Arginase 1 deficiency and their caregivers: A multinational, cross-sectional survey. JIMD Rep 2024; 65:450-460. [PMID: 39512431 PMCID: PMC11540579 DOI: 10.1002/jmd2.12456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 09/24/2024] [Accepted: 10/01/2024] [Indexed: 11/15/2024] Open
Abstract
Arginase 1 deficiency (ARG1-D) is an ultrarare, metabolic disease which may cause spastic paraplegia, cognitive deficiency, seizures, and ultimately severe disability. The aim of this study was to assess disease burden in ARG1-D by performing a cross-sectional survey of patients with ARG1-D and their caregivers in four European countries (France, Portugal, Spain, and the United Kingdom). Patients were enrolled at participating clinics and data were collected using a web-based questionnaire. The findings indicate that there is a significant share of patients who experience severe cognitive and mobility impairment but also that there is a considerable variance in symptom severity among patients. Disease management was mostly in line with treatment guidelines and self-reported adherence to treatment was reported to be high among a majority although following diet restrictions was perceived as difficult. However, despite this, since a large share of patients experienced severe cognitive and mobility impairment an unmet need among this patient population is suggested. The introduction of disease-modifying therapies and early identification and diagnosis may help alleviate the disease burden associated with ARG1-D in the future.
Collapse
Affiliation(s)
- Sara Olofsson
- The Swedish Institute for Health Economics, IHELundSweden
| | | | | | | | - Peter Lindgren
- The Swedish Institute for Health Economics, IHELundSweden
- Karolinska InstitutetStockholmSweden
| | - Karolina M. Stepien
- Salford Royal Organization, Northern Care Alliance NHS Foundation TrustSalfordUK
| | | | - Maria Luz Couce Pico
- Hospital Clinico Universitario de Santiago de Compostela, IDIS, MetabERNSantiago de CompostelaSpain
| | - Elisa Leão Teles
- Centro Hospitalar Universitário de São João, MetabERNPortoPortugal
| | | |
Collapse
|
19
|
Sarin Zacharia G, Jacob A. Ammonia in liver diseases: A glimpse into the controversies and consensus. Arab J Gastroenterol 2024; 25:323-329. [PMID: 39294031 DOI: 10.1016/j.ajg.2024.07.016] [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: 05/06/2024] [Revised: 06/20/2024] [Accepted: 07/26/2024] [Indexed: 09/20/2024]
Abstract
Ammonia is a byproduct of the metabolism of nitrogen-containing micro and macromolecules. The key source of bodily ammonia in humans is the small intestine, from diet, luminal bacterial activity, and deamination of glutamine in enterocytes. It is disposed of from the system, mainly in the liver, through the urea cycle. Physiologically, ammonia plays a minor role in acid-base homeostasis. It is the critical molecule implicated in the pathogenesis of hepatic encephalopathy. Liver disease is the most common cause of hyperammonemia, while others include urea cycle defects, infections, and drugs. The diagnostic utility of ammonia in liver diseases has met with increasing skepticism but holds good in urea cycle defects. Additionally, the accuracy of ammonia assay depends on a myriad of patient and technical factors, making the test unreliable if not performed meticulously. Most scientific societies currently fall short of recommending ammonia for diagnostic purposes in chronic liver disease. Despite this fact, ammonia continues to be one of the most frequently requested assays in patients with suspected hepatic encephalopathy, contributing to significant non-productive health expenditure. However, ammonia level does have a prognostic role in liver diseases, especially in acute liver failure. Ammonia-lowering strategies are the cornerstone of the management of hepatic encephalopathy. These strategies include medications that attenuate ammoniagenesis and ammonia scavengers. This review examines the role of ammonia in hepatic encephalopathy, its diagnostic and prognostic implications in liver diseases, challenges associated with ammonia assay, and current therapeutic strategies for ammonia-lowering in clinical practice.
Collapse
|
20
|
Caldovic L, Ahn JJ, Andricovic J, Balick VM, Brayer M, Chansky PA, Dawson T, Edwards AC, Felsen SE, Ismat K, Jagannathan SV, Mann BT, Medina JA, Morizono T, Morizono M, Salameh S, Vashist N, Williams EC, Zhou Z, Morizono H. Datamining approaches for examining the low prevalence of N-acetylglutamate synthase deficiency and understanding transcriptional regulation of urea cycle genes. J Inherit Metab Dis 2024; 47:1175-1193. [PMID: 37847851 PMCID: PMC11586597 DOI: 10.1002/jimd.12687] [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: 05/30/2023] [Revised: 10/09/2023] [Accepted: 10/10/2023] [Indexed: 10/19/2023]
Abstract
Ammonia, which is toxic to the brain, is converted into non-toxic urea, through a pathway of six enzymatically catalyzed steps known as the urea cycle. In this pathway, N-acetylglutamate synthase (NAGS, EC 2.3.1.1) catalyzes the formation of N-acetylglutamate (NAG) from glutamate and acetyl coenzyme A. NAGS deficiency (NAGSD) is the rarest of the urea cycle disorders, yet is unique in that ureagenesis can be restored with the drug N-carbamylglutamate (NCG). We investigated whether the rarity of NAGSD could be due to low sequence variation in the NAGS genomic region, high NAGS tolerance for amino acid replacements, and alternative sources of NAG and NCG in the body. We also evaluated whether the small genomic footprint of the NAGS catalytic domain might play a role. The small number of patients diagnosed with NAGSD could result from the absence of specific disease biomarkers and/or short NAGS catalytic domain. We screened for sequence variants in NAGS regulatory regions in patients suspected of having NAGSD and found a novel NAGS regulatory element in the first intron of the NAGS gene. We applied the same datamining approach to identify regulatory elements in the remaining urea cycle genes. In addition to the known promoters and enhancers of each gene, we identified several novel regulatory elements in their upstream regions and first introns. The identification of cis-regulatory elements of urea cycle genes and their associated transcription factors holds promise for uncovering shared mechanisms governing urea cycle gene expression and potentially leading to new treatments for urea cycle disorders.
Collapse
Affiliation(s)
- Ljubica Caldovic
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Julie J. Ahn
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Jacklyn Andricovic
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Veronica M. Balick
- Department of Biochemistry and Molecular MedicineThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
| | - Mallory Brayer
- Department of Biological SciencesThe George Washington UniversityWashingtonDCUSA
| | - Pamela A. Chansky
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Tyson Dawson
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- AMPEL BioSolutions LLCCharlottesvilleVirginiaUSA
| | - Alex C. Edwards
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Center for Neuroscience ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
| | - Sara E. Felsen
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Center for Neuroscience ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
| | - Karim Ismat
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Sveta V. Jagannathan
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Brendan T. Mann
- Department of Microbiology, Immunology, and Tropical MedicineSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Jacob A. Medina
- The Institute for Biomedical ScienceSchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
| | - Toshio Morizono
- College of Science and EngineeringUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Michio Morizono
- College of Science and EngineeringUniversity of MinnesotaMinneapolisMinnesotaUSA
| | - Shatha Salameh
- Department of Pharmacology & PhysiologySchool of Medicine and Health Sciences, George Washington UniversityWashingtonDCUSA
- Sheikh Zayed Institute for Pediatric Surgical InnovationChildren's National HospitalWashingtonDCUSA
| | - Neerja Vashist
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| | - Emily C. Williams
- Department of Anatomy and Cell BiologyThe George Washington University School of Medicine and Health SciencesWashingtonDCUSA
- The George Washington University Cancer Center, School of Medicine and Health SciencesGeorge Washington UniversityWashingtonDCUSA
| | - Zhe Zhou
- Department of Civil and Environmental EngineeringThe George Washington UniversityWashingtonDCUSA
| | - Hiroki Morizono
- Center for Genetic Medicine ResearchChildren's National Research Institute, Children's National HospitalWashingtonDCUSA
- Department of Genomics and Precision Medicine, School of Medicine and Health SciencesThe George Washington UniversityWashingtonDCUSA
| |
Collapse
|
21
|
Chanvanichtrakool M, Schreiber JM, Chen WL, Barber J, Zhang A, Ah Mew N, Schulze A, Wilkening G, Nagamani SCS, Gropman A. Unraveling the Link: Seizure Characteristics and Ammonia Levels in Urea Cycle Disorder During Hyperammonemic Crises. Pediatr Neurol 2024; 159:48-55. [PMID: 39121557 PMCID: PMC11381174 DOI: 10.1016/j.pediatrneurol.2024.06.013] [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: 03/30/2024] [Revised: 06/02/2024] [Accepted: 06/25/2024] [Indexed: 08/12/2024]
Abstract
BACKGROUND This retrospective clinical study performed at a single clinical center aimed to identify the prevalence of seizures in individuals with urea cycle disorders (UCDs) with and without hyperammonemic (HA) crises. In addition, we sought to correlate the utility of biochemical markers and electroencephalography (EEG) in detecting subclinical seizures during HA. METHODS Medical records of individuals with UCDs enrolled in Urea Cycle Disorders Consortium Longitudinal Study (UCDC-LS) (NCT00237315) at Children's National Hospital between 2006 and 2022 were reviewed for evidence of clinical and subclinical seizuress during HA crises, and initial biochemical levels concurrently. RESULTS Eighty-five individuals with UCD were included in the analyses. Fifty-six of the 85 patients (66%) experienced HA crises, with a total of 163 HA events. Seizures are observed in 13% of HA events. Among all HA events with concomitant EEG, subclinical seizures were identified in 27% of crises of encephalopathy without clinical seizures and 53% of crises with clinical seizures. The odds of seizures increases 2.65 (95% confidence interval [CI], 1.51 to 4.66) times for every 100 μmol/L increase in ammonia and 1.14 (95% CI, 1.04 to 1.25) times for every 100 μmol/L increase in glutamine. CONCLUSIONS This study highlights the utility of EEG monitoring during crises for patients presenting with clinical seizures or encephalopathy with HA. During HA events, measurement of initial ammonia and glutamine can help determine risk for seizures and guide EEG monitoring decisions.
Collapse
Affiliation(s)
- Mongkol Chanvanichtrakool
- Faculty of Medicine Siriraj Hospital, Division of Neurology, Department of Pediatrics, Mahidol University, Bangkok, Thailand; Division of Neurogenetics & Developmental Pediatrics, Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, District of Columbia
| | - John M Schreiber
- Division of Neurogenetics & Developmental Pediatrics, Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, District of Columbia
| | - Wei-Liang Chen
- Division of Neurogenetics & Developmental Pediatrics, Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, District of Columbia
| | - John Barber
- Division of Biostatistics and Study Methodology, Children's National Medical Center, Washington, District of Columbia
| | - Anqing Zhang
- Division of Biostatistics and Study Methodology, Children's National Medical Center, Washington, District of Columbia
| | - Nicholas Ah Mew
- Division of Genetics & Metabolism, Children's National Hospital, Washington, District of Columbia
| | - Andreas Schulze
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada; Departments of Pediatrics and Biochemistry, University of Toronto, Toronto, Canada
| | - Greta Wilkening
- Department of Pediatrics, Children's Hospital Colorado, University of Colorado, Aurora, Colorado
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Texas Children's Hospital, Houston, Texas
| | - Andrea Gropman
- Division of Neurogenetics & Developmental Pediatrics, Center for Neuroscience and Behavioral Medicine, Children's National Medical Center, Washington, District of Columbia.
| |
Collapse
|
22
|
Laochareonsuk W, Osatakul S, Intusoma U, Maneechay W, Sangkhathat S. A Novel Mutation of ORNT1 Detected in a Hyperornithinemia-Hyperammonemia-Homocitrullinuria Syndrome Child by Clinical Whole-Exome Sequencing. J Pediatr Genet 2024; 13:205-210. [PMID: 39086438 PMCID: PMC11288712 DOI: 10.1055/s-0041-1742247] [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: 08/24/2021] [Accepted: 12/21/2021] [Indexed: 10/19/2022]
Abstract
Hyperornithinemia-hyperammonemia-homocitrullinuria (HHH) syndrome, an inborn error of metabolism, is an inherited syndrome caused by loss-of-function mutations in the SLC25A15, resulting in ornithine translocase1 (ORNT1) deficiency. Disrupted ornithine transportation in an affected individual usually manifests with the accumulation of intermediate metabolites, leading to neurological impairment, hepatitis, and/or protein intolerance at various ages of onset. In this paper, we report a compound heterozygous mutation in SLC25A15 from a 2-year-old girl who presented with neurological alterations and hepatic failure. Before developing neurological sequelae, she had signs of globally delayed development. The accumulation of toxic metabolites may explain these neurological consequences. After biochemical confirmation of HHH, whole-exome sequencing (WES) was performed, which identified mutations at codons 21 and 179 of SLC25A15 that are predicted to result in the loss of function of ORNT1. Each of the mutations was found to be inherited from one of her parents. After therapy, her toxic metabolites decreased significantly. In conclusion, HHH syndrome frequently manifests with nonspecific symptoms and unapparent biochemical profiles, which may lead to delayed diagnosis. Correction of the accumulating metabolites is necessary to prevent irreversible neurological impairment. Furthermore, performing a WES provides a shortcut for accurate diagnosis.
Collapse
Affiliation(s)
- Wison Laochareonsuk
- Division of Biomedical Science and Biomedical Engineering, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Seksit Osatakul
- Pediatric Gastroenterology Unit, Division of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Utcharee Intusoma
- Pediatric Neurology Unit, Division of Pediatrics, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Wanwisa Maneechay
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| | - Surasak Sangkhathat
- Translational Medicine Research Center, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
- Division of Surgery, Faculty of Medicine, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
23
|
Zielonka M, Kölker S, Garbade SF, Gleich F, Nagamani SCS, Gropman AL, Druck AC, Ramdhouni N, Göde L, Hoffmann GF, Posset R. Severity-adjusted evaluation of initial dialysis on short-term health outcomes in urea cycle disorders. Mol Genet Metab 2024; 143:108566. [PMID: 39299137 DOI: 10.1016/j.ymgme.2024.108566] [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: 04/04/2024] [Revised: 08/07/2024] [Accepted: 08/13/2024] [Indexed: 09/22/2024]
Abstract
OBJECTIVE In individuals with urea cycle disorders (UCDs) and neonatal disease onset, extracorporeal detoxification by continuous kidney replacement therapy is considered the therapeutic method of choice in addition to metabolic emergency treatment to resolve hyperammonemic decompensation. However, the indications for the initiation of dialysis are heterogeneously implemented transnationally, thereby hampering our understanding of (optimal) short-term health outcomes. METHODS We performed a retrospective comparative analysis evaluating the therapeutic effects of initial dialysis on survival as well as neurocognitive outcome parameters in individuals with UCDs in comparison to a severity-adjusted non-dialyzed control cohort. Overall, 108 individuals with a severe phenotype of male ornithine transcarbamylase deficiency (mOTC-D), citrullinemia type 1 (CTLN1) and argininosuccinic aciduria (ASA) were investigated by stratification based on a recently established and validated genotype-specific disease prediction model. RESULTS Mortality is associated with the height of initial peak plasma ammonium concentration, but appears to be independent from treatment with initial dialysis in mOTC-D. However, improved survival after initial dialysis was observed in CTLN1, while there was a trend towards improved survival in ASA. In survivors, annual frequency of (subsequent) metabolic decompensations did not differ between the dialyzed and non-dialyzed cohorts. Moreover, treatment with initial dialysis was not associated with improved neurocognitive outcomes. INTERPRETATION The present severity-adjusted comparative analysis reveals that general practice of initial dialysis is neither associated with improved survival in individuals with mOTC-D nor does it differ with regard to the neurocognitive outcome for the investigated UCD subtypes. However, initial dialysis might potentially prove beneficial for survival in CTLN1 and ASA. CLINICAL TRIAL REGISTRATION The UCDC database is recorded at the US National Library of Medicine (https://clinicaltrials.gov).
Collapse
Affiliation(s)
- Matthias Zielonka
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Stefan Kölker
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Sven F Garbade
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Florian Gleich
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Andrea L Gropman
- Children's National Health System and The George Washington School of Medicine, Washington, DC, USA
| | - Ann-Catrin Druck
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Nesrine Ramdhouni
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Laura Göde
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Georg F Hoffmann
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Roland Posset
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| |
Collapse
|
24
|
Tran DM, Tran TTT, Luong QH, Tran MTC. A preliminary retrospective evaluation of screening and diagnosis of ornithine transcarbamylase deficiency in high-risk patients at a referral center in Vietnam. Heliyon 2024; 10:e36003. [PMID: 39220945 PMCID: PMC11365392 DOI: 10.1016/j.heliyon.2024.e36003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 08/04/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Introduction To date, newborn screening (NBS) for proximal urea cycle disorders, including Ornithine transcarbamylase deficiency (OTCD), was not recommended due to the lack of appropriate tests and insufficient evidence of the benefits. This study aimed to investigate the potential of tandem mass spectrometry (MS/MS) for OTCD screening and its value in guiding further investigation to obtain a final diagnosis in high-risk patients. Methods The study included patients with OTCD referred to the National Children's Hospital between April 2020 and November 2023. A retrospective evaluation of amino acid concentrations measured by MS/MS and their ratios in patients with early-onset and late-onset OTCD was conducted. Results While all ten early-onset cases had glutamine concentrations above the upper limit, only five of them had citrulline concentrations below the lower limit of the reference interval. Only two late-onset cases had elevated glutamine levels, while all had citrulline within reference intervals. The Cit/Phe ratio was decreased, and the Gln/Cit and Met/Cit ratios were increased in all early-onset OTCD cases, while they were abnormal in only one late-onset case. Conclusions The preliminary results suggest that hyperglutaminemia, in combination with low or normal citrulline concentrations and specific ratios (Gln/Cit, Met/Cit, and Cit/Phe), can serve as reliable markers for screening early-onset OTCD in high-risk patients. However, these markers proved less sensitive for detecting the late-onset form, even in symptomatic patients.
Collapse
Affiliation(s)
| | | | | | - Mai Thi Chi Tran
- National Children's Hospital, Hanoi, Viet Nam
- Hanoi Medical University, Hanoi, Viet Nam
| |
Collapse
|
25
|
Dong H, Sang T, Ma X, Song J, Chen Z, Zhang H, Jin Y, Li M, Dong D, Sun L, Zhu Z, Zhang Y, Yang Y. Clinical features and CPS1 variants in Chinese patients with carbamoyl phosphate synthetase 1 deficiency. BMC Pediatr 2024; 24:539. [PMID: 39174957 PMCID: PMC11340094 DOI: 10.1186/s12887-024-05005-5] [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: 01/28/2024] [Accepted: 08/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Carbamoyl phosphate synthetase 1 (CPS1) deficiency (OMIM 237300), an autosomal recessive rare and severe urea cycle disorder, is associated with hyperammonemia and high mortality. METHODS Herein we present 12 genetic variants identified in seven clinically well-characterized Chinese patients with CPS1 deficiency who were admitted to the Children's Medical Center of Peking University First Hospital from September 2014 to August 2023. RESULTS Seven patients (two male and five female patients including two sisters) experienced symptoms onset between 2 days and 13 years of age, and they were diagnosed with CPS1 deficiency between 2 months and 20 years. Peak blood ammonia levels ranged from 160 to 1,000 µmol/L. Three patients showed early-onset CPS1 deficiency, with only one surviving after treatment with sodium phenylbutyrate, N-carbamoyl-L-glutamate, and liver transplantation at 4 months, showing a favorable outcome. The remaining four patients had late-onset CPS1 deficiency, presenting with mental retardation, psychiatric symptoms, and self-selected low-protein diets. Among the 12 CPS1 variants identified in these patients, 10 were novel, with all patients exhibiting compound heterozygosity for CPS1 mutant alleles. Seven variants (c.149T > C, c.616 A > T, c.1145 C > T, c.1294G > A, c.3029 C > T, c.3503 A > T, and c.3793 C > T) resulted in single amino acid substitutions. Three frameshift variations (c.2493del, c.3067dup, and c.3241del) were identified, leading to enzyme truncation. One mutation (c.3506_3508del) caused an in-frame single amino acid deletion, while another (c.2895 + 2T > C) resulted in aberrant splicing. CONCLUSIONS Except for two known variants, all other variants were identified as novel. No hotspot variants were observed among the patients. Our data contribute to expanding the mutation spectrum of CPS1.
Collapse
Affiliation(s)
- Hui Dong
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Tian Sang
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Xue Ma
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Jinqing Song
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Zhehui Chen
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
- Department of Pediatrics, Women and Children's Hospital, School of Medicine, Xiamen University, Xiamen, 361003, China
| | - Huiting Zhang
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Ying Jin
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Mengqiu Li
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Dingding Dong
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China
| | - Liying Sun
- Department of General Surgery, Beijing Friendship Hospital of Capital Medical University, Beijing, 100050, China
| | - Zhijun Zhu
- Department of General Surgery, Beijing Friendship Hospital of Capital Medical University, Beijing, 100050, China
| | - Yao Zhang
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China.
| | - Yanling Yang
- Children's Medical Center, Peking University First Hospital, Beijing, 102600, China.
| |
Collapse
|
26
|
Rodríguez-Pombo L, de Castro-López MJ, Sánchez-Pintos P, Giraldez-Montero JM, Januskaite P, Duran-Piñeiro G, Dolores Bóveda M, Alvarez-Lorenzo C, Basit AW, Goyanes A, Couce ML. Paediatric clinical study of 3D printed personalised medicines for rare metabolic disorders. Int J Pharm 2024; 657:124140. [PMID: 38643809 DOI: 10.1016/j.ijpharm.2024.124140] [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: 03/08/2024] [Revised: 04/17/2024] [Accepted: 04/18/2024] [Indexed: 04/23/2024]
Abstract
Rare diseases are infrequent, but together they affect up to 6-10 % of the world's population, mainly children. Patients require precise doses and strict adherence to avoid metabolic or cardiac failure in some cases, which cannot be addressed in a reliable way using pharmaceutical compounding. 3D printing (3DP) is a disruptive technology that allows the real-time personalization of the dose and the modulation of the dosage form to adapt the medicine to the therapeutic needs of each patient. 3D printed chewable medicines containing amino acids (citrulline, isoleucine, valine, and isoleucine and valine combinations) were prepared in a hospital setting, and the efficacy and acceptability were evaluated in comparison to conventional compounded medicines in six children. The inclusion of new flavours (lemon, vanilla and peach) to obtain more information on patient preferences and the implementation of a mobile app to obtain patient feedback in real-time was also used. The 3D printed medicines controlled amino acid levels within target levels as well as the conventional medicines. The deviation of citrulline levels was narrower and closer within the target concentration with the chewable formulations. According to participants' responses, the chewable formulations were well accepted and can improve adherence and quality of life. For the first time, 3DP enabled two actives to be combined in the same formulation, reducing the number of administrations. This study demonstrated the benefits of preparing 3D printed personalized treatments for children diagnosed with rare metabolic disorders using a novel technology in real clinical practice.
Collapse
Affiliation(s)
- Lucía Rodríguez-Pombo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Materials Institute iMATUS and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - María José de Castro-López
- Servicio de Neonatología, Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Universidad de Santiago de Compostela, IDIS, RICORS, CIBERER, MetabERN, Spain
| | - Paula Sánchez-Pintos
- Servicio de Neonatología, Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Universidad de Santiago de Compostela, IDIS, RICORS, CIBERER, MetabERN, Spain
| | - Jose Maria Giraldez-Montero
- Pharmacy Department, Xerencia de Xestión Integrada de Santiago de Compostela, SERGAS, Travesía Choupana s/n, Santiago de Compostela 15706, Spain
| | - Patricija Januskaite
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK
| | - Goretti Duran-Piñeiro
- Pharmacy Department, Xerencia de Xestión Integrada de Santiago de Compostela, SERGAS, Travesía Choupana s/n, Santiago de Compostela 15706, Spain
| | - M Dolores Bóveda
- Servicio de Neonatología, Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Universidad de Santiago de Compostela, IDIS, RICORS, CIBERER, MetabERN, Spain
| | - Carmen Alvarez-Lorenzo
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Materials Institute iMATUS and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain
| | - Abdul W Basit
- Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; FABRX Ltd., Henwood House, Henwood, Ashford, Kent TN24 8DH, UK; FABRX Artificial Intelligence, Carretera de Escairón, 14, Currelos (O Saviñao), CP 27543, Spain
| | - Alvaro Goyanes
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, I+D Farma (GI-1645), Facultad de Farmacia, Materials Institute iMATUS and Health Research Institute of Santiago de Compostela (IDIS), Universidade de Santiago de Compostela, 15782 Santiago de Compostela, Spain; Department of Pharmaceutics, UCL School of Pharmacy, University College London, 29-39 Brunswick Square, London WC1N 1AX, UK; FABRX Ltd., Henwood House, Henwood, Ashford, Kent TN24 8DH, UK; FABRX Artificial Intelligence, Carretera de Escairón, 14, Currelos (O Saviñao), CP 27543, Spain.
| | - Maria L Couce
- Servicio de Neonatología, Unidad de Diagnóstico y Tratamiento de Enfermedades Metabólicas Congénitas, Health Research Institute of Santiago de Compostela (IDIS), Hospital Clínico Universitario de Santiago de Compostela, Universidad de Santiago de Compostela, IDIS, RICORS, CIBERER, MetabERN, Spain.
| |
Collapse
|
27
|
杨 帆, 王 立, 李 辛, 胡 佳, 应 令, 冯 碧, 李 芸, 林 卡, 佘 佳, 李 浩, 常 国, 王 秀. [Treatment of ornithine transcarbamylase deficiency in a child with glyceryl phenylbutyrate]. ZHONGGUO DANG DAI ER KE ZA ZHI = CHINESE JOURNAL OF CONTEMPORARY PEDIATRICS 2024; 26:512-517. [PMID: 38802913 PMCID: PMC11135055 DOI: 10.7499/j.issn.1008-8830.2310050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 02/19/2024] [Indexed: 05/29/2024]
Abstract
Glyceryl phenylbutyrate (GPB) serves as a long-term management medication for Ornithine transcarbamylase deficiency (OTCD), effectively controlling hyperammonemia, but there is a lack of experience in using this medicine in China. This article retrospectively analyzes the case of a child diagnosed with OTCD at Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, including a review of related literature. After diagnosis, the patient was treated with GPB, followed by efficacy follow-up and pharmacological monitoring. The 6-year and 6-month-old male patient exhibited poor speech development, disobedience, temper tantrums, and aggressive behavior. Blood ammonia levels peaked at 327 μmol/L; urine organic acid analysis indicated elevated uracil levels; cranial MRI showed extensive abnormal signals in both cerebral hemispheres. Genetic testing revealed de novo mutation in the OTC gene (c.241T>C, p.S81P). Blood ammonia levels were approximately 43, 80, and 56 μmol/L at 1, 2, and 3 months after starting GPB treatment, respectively. During treatment, blood ammonia was well-controlled without drug-related adverse effects. The patient showed improvement in developmental delays, obedience, temperament, and absence of aggressive behavior.
Collapse
Affiliation(s)
| | - 立瑞 王
- 上海交通大学医学院附属上海儿童医学中心内分泌遗传代谢科上海200127
- 云南省德宏州陇川县人民医院儿科,云南德宏678700
| | - 辛 李
- 上海交通大学医学院附属上海儿童医学中心内分泌遗传代谢科上海200127
| | | | - 令雯 应
- 上海交通大学医学院附属上海儿童医学中心内分泌遗传代谢科上海200127
| | - 碧云 冯
- 上海交通大学医学院附属上海儿童医学中心内分泌遗传代谢科上海200127
| | | | | | | | | | - 国营 常
- 上海交通大学医学院附属上海儿童医学中心内分泌遗传代谢科上海200127
| | - 秀敏 王
- 上海交通大学医学院附属上海儿童医学中心内分泌遗传代谢科上海200127
| |
Collapse
|
28
|
Singh RH, Bourdages MH, Kurtz A, MacLoed E, Norman C, Ratko S, van Calcar SC, Kenneson A. The efficacy of Carbamylglutamate impacts the nutritional management of patients with N-Acetylglutamate synthase deficiency. Orphanet J Rare Dis 2024; 19:168. [PMID: 38637895 PMCID: PMC11027358 DOI: 10.1186/s13023-024-03167-0] [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: 11/08/2023] [Accepted: 03/30/2024] [Indexed: 04/20/2024] Open
Abstract
BACKGROUND The autosomal recessive disorder N-acetylglutamate synthase (NAGS) deficiency is the rarest defect of the urea cycle, with an incidence of less than one in 2,000,000 live births. Hyperammonemic crises can be avoided in individuals with NAGS deficiency by the administration of carbamylglutamate (also known as carglumic acid), which activates carbamoyl phosphatase synthetase 1 (CPS1). The aim of this case series was to introduce additional cases of NAGS deficiency to the literature as well as to assess the role of nutrition management in conjunction with carbamylglutamate therapy across new and existing cases. METHODS We conducted retrospective chart reviews of seven cases of NAGS deficiency in the US and Canada, focusing on presentation, diagnosis, medication management, nutrition management, and outcomes. RESULTS Five new and two previously published cases were included. Presenting symptoms were consistent with previous reports. Diagnostic confirmation via molecular testing varied in protocol across cases, with consecutive single gene tests leading to long delays in diagnosis in some cases. All patients responded well to carbamylglutamate therapy, as indicated by normalization of plasma ammonia and citrulline, as well as urine orotic acid in patients with abnormal levels at baseline. Although protein restriction was not prescribed in any cases after carbamylglutamate initiation, two patients continued to self-restrict protein intake. One patient experienced two episodes of hyperammonemia that resulted in poor long-term outcomes. Both episodes occurred after a disruption in access to carbamylglutamate, once due to insurance prior authorization requirements and language barriers and once due to seizure activity limiting the family's ability to administer carbamylglutamate. CONCLUSIONS Follow-up of patients with NAGS deficiency should include plans for illness and for disruption of carbamylglutamate access, including nutrition management strategies such as protein restriction. Carbamylglutamate can help patients with NAGS deficiency to liberalize their diets, but the maximum safe level of protein intake to prevent hyperammonemia is not yet known. Patients using this medication should still monitor their diet closely and be prepared for any disruptions in medication access, which might require immediate dietary adjustments or medical intervention to prevent hyperammonemia.
Collapse
Affiliation(s)
- Rani H Singh
- Emory University School of Medicine, 101 Woodruff Circle, 7th Floor Suite 7130, 30322, Atlanta, GA, USA.
| | | | | | - Erin MacLoed
- Children's National Medical Center, Washington, DC, USA
| | | | | | | | - Aileen Kenneson
- Emory University School of Medicine, 101 Woodruff Circle, 7th Floor Suite 7130, 30322, Atlanta, GA, USA
| |
Collapse
|
29
|
Jalil S, Keskinen T, Juutila J, Sartori Maldonado R, Euro L, Suomalainen A, Lapatto R, Kuuluvainen E, Hietakangas V, Otonkoski T, Hyvönen ME, Wartiovaara K. Genetic and functional correction of argininosuccinate lyase deficiency using CRISPR adenine base editors. Am J Hum Genet 2024; 111:714-728. [PMID: 38579669 PMCID: PMC11023919 DOI: 10.1016/j.ajhg.2024.03.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 03/06/2024] [Accepted: 03/06/2024] [Indexed: 04/07/2024] Open
Abstract
Argininosuccinate lyase deficiency (ASLD) is a recessive metabolic disorder caused by variants in ASL. In an essential step in urea synthesis, ASL breaks down argininosuccinate (ASA), a pathognomonic ASLD biomarker. The severe disease forms lead to hyperammonemia, neurological injury, and even early death. The current treatments are unsatisfactory, involving a strict low-protein diet, arginine supplementation, nitrogen scavenging, and in some cases, liver transplantation. An unmet need exists for improved, efficient therapies. Here, we show the potential of a lipid nanoparticle-mediated CRISPR approach using adenine base editors (ABEs) for ASLD treatment. To model ASLD, we first generated human-induced pluripotent stem cells (hiPSCs) from biopsies of individuals homozygous for the Finnish founder variant (c.1153C>T [p.Arg385Cys]) and edited this variant using the ABE. We then differentiated the hiPSCs into hepatocyte-like cells that showed a 1,000-fold decrease in ASA levels compared to those of isogenic non-edited cells. Lastly, we tested three different FDA-approved lipid nanoparticle formulations to deliver the ABE-encoding RNA and the sgRNA targeting the ASL variant. This approach efficiently edited the ASL variant in fibroblasts with no apparent cell toxicity and minimal off-target effects. Further, the treatment resulted in a significant decrease in ASA, to levels of healthy donors, indicating restoration of the urea cycle. Our work describes a highly efficient approach to editing the disease-causing ASL variant and restoring the function of the urea cycle. This method relies on RNA delivered by lipid nanoparticles, which is compatible with clinical applications, improves its safety profile, and allows for scalable production.
Collapse
Affiliation(s)
- Sami Jalil
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Timo Keskinen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Juhana Juutila
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Rocio Sartori Maldonado
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Liliya Euro
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Anu Suomalainen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Risto Lapatto
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Emilia Kuuluvainen
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Ville Hietakangas
- Faculty of Biological and Environmental Sciences University of Helsinki, Helsinki, Finland; Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Timo Otonkoski
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Mervi E Hyvönen
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; New Children's Hospital, Pediatric Research Center, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | - Kirmo Wartiovaara
- Stem Cells and Metabolism Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland; Clinical Genetics, Helsinki University Hospital, Helsinki, Finland.
| |
Collapse
|
30
|
Moedas MF, Simões RJM, Silva MFB. Mitochondrial targets in hyperammonemia: Addressing urea cycle function to improve drug therapies. Biochem Pharmacol 2024; 222:116034. [PMID: 38307136 DOI: 10.1016/j.bcp.2024.116034] [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: 10/28/2023] [Revised: 12/27/2023] [Accepted: 01/25/2024] [Indexed: 02/04/2024]
Abstract
The urea cycle (UC) is a critically important metabolic process for the disposal of nitrogen (ammonia) produced by amino acids catabolism. The impairment of this liver-specific pathway induced either by primary genetic defects or by secondary causes, namely those associated with hepatic disease or drug administration, may result in serious clinical consequences. Urea cycle disorders (UCD) and certain organic acidurias are the major groups of inherited rare diseases manifested with hyperammonemia (HA) with UC dysregulation. Importantly, several commonly prescribed drugs, including antiepileptics in monotherapy or polytherapy from carbamazepine to valproic acid or specific antineoplastic agents such as asparaginase or 5-fluorouracil may be associated with HA by mechanisms not fully elucidated. HA, disclosing an imbalance between ammoniagenesis and ammonia disposal via the UC, can evolve to encephalopathy which may lead to significant morbidity and central nervous system damage. This review will focus on biochemical mechanisms related with HA emphasizing some poorly understood perspectives behind the disruption of the UC and mitochondrial energy metabolism, namely: i) changes in acetyl-CoA or NAD+ levels in subcellular compartments; ii) post-translational modifications of key UC-related enzymes, namely acetylation, potentially affecting their catalytic activity; iii) the mitochondrial sirtuins-mediated role in ureagenesis. Moreover, the main UCD associated with HA will be summarized to highlight the relevance of investigating possible genetic mutations to account for unexpected HA during certain pharmacological therapies. The ammonia-induced effects should be avoided or overcome as part of safer therapeutic strategies to protect patients under treatment with drugs that may be potentially associated with HA.
Collapse
Affiliation(s)
- Marco F Moedas
- Research Institute for Medicines-iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal; Centre for Inherited Metabolic Diseases, Karolinska University Hospital, Stockholm, Sweden; Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Ricardo J M Simões
- Research Institute for Medicines-iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal
| | - Margarida F B Silva
- Research Institute for Medicines-iMed.ULisboa, Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003 Lisboa, Portugal.
| |
Collapse
|
31
|
Sen K, Izem R, Long Y, Jiang J, Konczal LL, McCarter RJ, Gropman AL, Bedoyan JK. Are asymptomatic carriers of OTC deficiency always asymptomatic? A multicentric retrospective study of risk using the UCDC longitudinal study database. Mol Genet Genomic Med 2024; 12:e2443. [PMID: 38634223 PMCID: PMC11024633 DOI: 10.1002/mgg3.2443] [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: 11/18/2023] [Revised: 03/24/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
BACKGROUND Ornithine transcarbamylase deficiency (OTCD) due to an X-linked OTC mutation, is responsible for moderate to severe hyperammonemia (HA) with substantial morbidity and mortality. About 80% of females with OTCD remain apparently "asymptomatic" with limited studies of their clinical characteristics and long-term health vulnerabilities. Multimodal neuroimaging studies and executive function testing have shown that asymptomatic females exhibit limitations when stressed to perform at higher cognitive load and had reduced activation of the prefrontal cortex. This retrospective study aims to improve understanding of factors that might predict development of defined complications and serious illness in apparent asymptomatic females. A proband and her daughter are presented to highlight the utility of multimodal neuroimaging studies and to underscore that asymptomatic females with OTCD are not always asymptomatic. METHODS We review data from 302 heterozygote females with OTCD enrolled in the Urea Cycle Disorders Consortium (UCDC) longitudinal natural history database. We apply multiple neuroimaging modalities in the workup of a proband and her daughter. RESULTS Among the females in the database, 143 were noted as symptomatic at baseline (Sym). We focused on females who were asymptomatic (Asx, n = 111) and those who were asymptomatic initially upon enrollment in study but who later became symptomatic sometime during follow-up (Asx/Sym, n = 22). The majority of Asx (86%) and Asx/Sym (75%) subjects did not restrict protein at baseline, and ~38% of Asx and 33% of Asx/Sym subjects suffered from mild to severe neuropsychiatric conditions such as mood disorder and sleep problems. The risk of mild to severe HA sometime later in life for the Asx and Asx/Sym subjects as a combined group was ~4% (5/133), with ammonia ranging from 77 to 470 μM and at least half (2/4) of subjects requiring hospital admission and nitrogen scavenger therapy. For this combined group, the median age of first HA crisis was 50 years, whereas the median age of first symptom which included neuropsychiatric and/or behavioral symptoms was 17 years. The multimodal neuroimaging studies in female heterozygotes with OTCD also underscore that asymptomatic female heterozygotes with OTCD (e.g., proband) are not always asymptomatic. CONCLUSIONS Analysis of Asx and Asx/Sym females with OTCD in this study suggests that future evidence-based management guidelines and/or a clinical risk score calculator for this cohort could be useful management tools to reduce morbidity and improve long-term quality of life.
Collapse
Affiliation(s)
- Kuntal Sen
- Division of Neurogenetics and Neurodevelopmental PediatricsChildren's National Hospital, The George Washington School of MedicineWashingtonDCUSA
| | - Rima Izem
- Center for Translational Sciences, Children's National HospitalThe George Washington UniversityWashingtonDCUSA
- Children's National HospitalWashingtonDCUSA
| | - Yuelin Long
- Columbia University Mailman School of Public HealthNew YorkNew YorkUSA
| | - Jiji Jiang
- Center for Translational Sciences, Children's National HospitalThe George Washington UniversityWashingtonDCUSA
- Children's National HospitalWashingtonDCUSA
| | - Laura L. Konczal
- Center for Human Genetics, University Hospitals Cleveland Medical Center, Department of Genetics and Genome SciencesCase Western Reserve University School of MedicineClevelandOhioUSA
| | - Robert J. McCarter
- Center for Translational Sciences, Children's National HospitalThe George Washington UniversityWashingtonDCUSA
- Children's National HospitalWashingtonDCUSA
| | - Andrea L. Gropman
- Division of Neurogenetics and Neurodevelopmental PediatricsChildren's National Hospital, The George Washington School of MedicineWashingtonDCUSA
- Center for Translational Sciences, Children's National HospitalThe George Washington UniversityWashingtonDCUSA
| | - Jirair K. Bedoyan
- Division of Genetic and Genomic Medicine, Department of PediatricsUPMC Children's Hospital of Pittsburgh, University of Pittsburgh School of MedicinePittsburghPennsylvaniaUSA
| |
Collapse
|
32
|
Posset R, Garbade SF, Gleich F, Nagamani SCS, Gropman AL, Epp F, Ramdhouni N, Druck AC, Hoffmann GF, Kölker S, Zielonka M. Impact of supplementation with L-citrulline/arginine after liver transplantation in individuals with Urea Cycle Disorders. Mol Genet Metab 2024; 141:108112. [PMID: 38301530 DOI: 10.1016/j.ymgme.2023.108112] [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: 10/25/2023] [Revised: 12/06/2023] [Accepted: 12/06/2023] [Indexed: 02/03/2024]
Abstract
OBJECTIVE Liver transplantation (LTx) is an intervention when medical management is not sufficiently preventing individuals with urea cycle disorders (UCDs) from the occurrence of hyperammonemic events. Supplementation with L-citrulline/arginine is regularly performed prior to LTx to support ureagenesis and is often continued after the intervention. However, systematic studies assessing the impact of long-term L-citrulline/arginine supplementation in individuals who have undergone LTx is lacking to date. METHODS Using longitudinal data collected systematically, a comparative analysis was carried out by studying the effects of long-term L-citrulline/arginine supplementation vs. no supplementation on health-related outcome parameters (i.e., anthropometric, neurological, and cognitive outcomes) in individuals with UCDs who have undergone LTx. Altogether, 52 individuals with male ornithine transcarbamylase deficiency, citrullinemia type 1 and argininosuccinic aciduria and a pre-transplant "severe" disease course who have undergone LTx were investigated by using recently established and validated genotype-specific in vitro enzyme activities. RESULTS Long-term supplementation of individuals with L-citrulline/arginine who have undergone LTx (n = 16) does neither appear to alter anthropometric nor neurocognitive endpoints when compared to their severity-adjusted counterparts that were not supplemented (n = 36) after LTx with mean observation periods between four to five years. Moreover, supplementation with L-citrulline/arginine was not associated with an increase of disease-specific plasma arithmetic mean values for the respective amino acids when compared to the non-supplemented control cohort. CONCLUSION Although supplementation with L-citrulline/arginine is often continued after LTx, this pilot study does neither identify altered long-term anthropometric or neurocognitive health-related outcomes nor does it find an adequate biochemical response as reflected by the unaltered plasma arithmetic mean values for L-citrulline or L-arginine. Further prospective analyses in larger samples and even longer observation periods will provide more insight into the usefulness of long-term supplementation with L-citrulline/arginine for individuals with UCDs who have undergone LTx.
Collapse
Affiliation(s)
- Roland Posset
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| | - Sven F Garbade
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Florian Gleich
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine and Texas Children's Hospital, Houston, TX, USA
| | - Andrea L Gropman
- Children's National Health System and The George Washington School of Medicine, Washington, DC, USA
| | - Friederike Epp
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Nesrine Ramdhouni
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Ann-Catrin Druck
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Georg F Hoffmann
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Stefan Kölker
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany
| | - Matthias Zielonka
- Heidelberg University, Medical Faculty Heidelberg, and Center for Pediatric and Adolescent Medicine, Department I, Division of Pediatric Neurology and Metabolic Medicine, University Hospital Heidelberg, Im Neuenheimer Feld 430, 69120 Heidelberg, Germany.
| |
Collapse
|
33
|
Cameron JM, Osundiji MA, Olson RJ, Olarewaju BA, Schulze A. ACMG/AMP variant classification framework in arginase 1 deficiency: Implications for birth prevalence estimates and diagnostics. GENETICS IN MEDICINE OPEN 2024; 2:101815. [PMID: 39669610 PMCID: PMC11613747 DOI: 10.1016/j.gimo.2024.101815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/04/2023] [Revised: 01/15/2024] [Accepted: 01/18/2024] [Indexed: 12/14/2024]
Abstract
Purpose Arginase 1 (ARG1) deficiency manifests with hyperargininemia and progressive neurological impairment. Recent estimates of birth prevalence using allele frequencies of ARG1 variants do not sufficiently distinguish benign from pathogenic variants. Additionally, ongoing discussions of reproductive carrier screening for diseases such as ARG1 creates a need for improved understanding of ARG1 variant classification. Here, we incorporate American College of Medical Genetics and Genomics/Association for Molecular Pathology-developed guidelines for interpreting gene variants and in silico predictions to select allele frequencies for estimation of global birth prevalence of ARG1 deficiency. Methods We interrogated Genome Aggregation Database and PubMed for published (defined as identified in patients with clinically defined arginase deficiency in scientific literature, n = 73) and unpublished ARG1 variants (defined as variants present in Genome Aggregation Database, unique to ARG1, but not yet associated with clinical arginase deficiency, n = 302). American College of Medical Genetics and Genomics/Association for Molecular Pathology guidelines were applied to classify variants using Franklin Genoox artificial intelligence-powered platform and manual review. Results Of 73 published ARG1 variants, 16 classified as pathogenic, 30 as likely pathogenic, and 27 as variant of uncertain significance. Of 302 unpublished ARG1 variants, 3 classified as pathogenic, 28 likely pathogenic, and 229 variant of uncertain significance. Mutant allele frequency estimates ranged from 17 to 266 per 100,000 and birth prevalence from 1 in 141,331 to 34,602,076. Conclusion We show that a large proportion of ARG1 variants lack adequate evidence of pathogenicity. These findings underscore the significance of functional studies and accumulating clinical data for determination of variant pathogenicity and for improved understanding of global birth prevalence of ARG1 deficiency.
Collapse
Affiliation(s)
- Jessie M. Cameron
- Department of Paediatric Laboratory Medicine, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Mayowa Azeez Osundiji
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Rory J. Olson
- Department of Clinical Genomics, Mayo Clinic, Rochester, MN
| | - Bukola A. Olarewaju
- School of Science and Engineering, University of Dundee, Dundee, United Kingdom
| | - Andreas Schulze
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
34
|
Gurung S, Timmermand OV, Perocheau D, Gil-Martinez AL, Minnion M, Touramanidou L, Fang S, Messina M, Khalil Y, Spiewak J, Barber AR, Edwards RS, Pinto PL, Finn PF, Cavedon A, Siddiqui S, Rice L, Martini PGV, Ridout D, Heywood W, Hargreaves I, Heales S, Mills PB, Waddington SN, Gissen P, Eaton S, Ryten M, Feelisch M, Frassetto A, Witney TH, Baruteau J. mRNA therapy corrects defective glutathione metabolism and restores ureagenesis in preclinical argininosuccinic aciduria. Sci Transl Med 2024; 16:eadh1334. [PMID: 38198573 PMCID: PMC7615535 DOI: 10.1126/scitranslmed.adh1334] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2023] [Accepted: 10/06/2023] [Indexed: 01/12/2024]
Abstract
The urea cycle enzyme argininosuccinate lyase (ASL) enables the clearance of neurotoxic ammonia and the biosynthesis of arginine. Patients with ASL deficiency present with argininosuccinic aciduria, an inherited metabolic disease with hyperammonemia and a systemic phenotype coinciding with neurocognitive impairment and chronic liver disease. Here, we describe the dysregulation of glutathione biosynthesis and upstream cysteine utilization in ASL-deficient patients and mice using targeted metabolomics and in vivo positron emission tomography (PET) imaging using (S)-4-(3-18F-fluoropropyl)-l-glutamate ([18F]FSPG). Up-regulation of cysteine metabolism contrasted with glutathione depletion and down-regulated antioxidant pathways. To assess hepatic glutathione dysregulation and liver disease, we present [18F]FSPG PET as a noninvasive diagnostic tool to monitor therapeutic response in argininosuccinic aciduria. Human hASL mRNA encapsulated in lipid nanoparticles improved glutathione metabolism and chronic liver disease. In addition, hASL mRNA therapy corrected and rescued the neonatal and adult Asl-deficient mouse phenotypes, respectively, enhancing ureagenesis. These findings provide mechanistic insights in liver glutathione metabolism and support clinical translation of mRNA therapy for argininosuccinic aciduria.
Collapse
Affiliation(s)
- Sonam Gurung
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | | | - Dany Perocheau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ana Luisa Gil-Martinez
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Magdalena Minnion
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | - Loukia Touramanidou
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Sherry Fang
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Martina Messina
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Youssef Khalil
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Justyna Spiewak
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Abigail R Barber
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Richard S Edwards
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Patricia Lipari Pinto
- Santa Maria's Hospital, Lisbon North University Hospital Center, 1649-028 Lisbon, Portugal
| | | | | | | | - Lisa Rice
- Moderna Inc., Cambridge, MA 02139, USA
| | | | - Deborah Ridout
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Wendy Heywood
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Ian Hargreaves
- Pharmacy and Biomolecular Sciences, Liverpool John Moore University, Liverpool L3 5UG, UK
| | - Simon Heales
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Philippa B Mills
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Simon N Waddington
- EGA Institute for Women's Health, University College London, London WC1E 6HX, UK
- Wits/SAMRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of Witswatersrand, Braamfontein, 2000 Johannesburg, South Africa
| | - Paul Gissen
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| | - Simon Eaton
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Mina Ryten
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Martin Feelisch
- Clinical and Experimental Sciences, Faculty of Medicine, University of Southampton, Southampton SO17 1BJ, UK
- Southampton NIHR Biomedical Research Centre, University Hospital Southampton NHS Foundation Trust, Southampton SO16 6YD, UK
| | | | - Timothy H Witney
- School of Biomedical Engineering and Imaging Sciences, King's College London, London SE1 7EH, UK
| | - Julien Baruteau
- Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
- National Institute of Health Research Great Ormond Street Biomedical Research Centre, London WC1N 1EH, UK
| |
Collapse
|
35
|
Kakiuchi T, Nosho T, Oka M, Tashiro K. Hyperammonemia in a carbamoyl-phosphate synthetase 1 deficiency recipient after living-donor liver transplantation from a carrier donor: a case report. Front Med (Lausanne) 2024; 10:1327854. [PMID: 38235270 PMCID: PMC10792046 DOI: 10.3389/fmed.2023.1327854] [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: 10/25/2023] [Accepted: 12/13/2023] [Indexed: 01/19/2024] Open
Abstract
Carbamoyl-phosphate synthetase 1 (CPS1) deficiency is an autosomal recessive congenital urea cycle disorder (UCD) characterized by hyperammonemia. The recipients of liver transplantation (LT) for UCD are often children, and the potential donors are often the parents. Hereditary congenital diseases involving UCD entail the possibility of both parents being genetically heterozygous. Herein, we describe the case of a 12-year-old girl with CPS1 deficiency receiving a liver transplant (soon after birth) from her father, who had a heterozygous CPS1 mutation. She was referred to our hospital with respiratory distress after contracting two infections (respiratory syncytial virus and human metapneumovirus) within a short period, both of which presented with hyperammonemia. Medication for hyperammonemia quickly lowered the ammonia levels. The hyperammonemia was thought to be caused by the heterozygous mutation in the donor liver; moreover, it is likely that the low enzyme activity in the patient's liver was increased due to the infections. This is the first study to report hyperammonemia in a CPS1 deficiency patient due to an infection after LT. Thus, patients with CPS1 deficiency should be aware of the development of hyperammonemia after LT.
Collapse
Affiliation(s)
- Toshihiko Kakiuchi
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Tetsuya Nosho
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Masafumi Oka
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Katsuya Tashiro
- Department of Pediatrics, Karatsu Red Cross Hospital, Karatsu, Japan
| |
Collapse
|
36
|
Siri B, Olivieri G, Lepri FR, Poms M, Goffredo BM, Commone A, Novelli A, Häberle J, Dionisi-Vici C. Father-to-daughter transmission in late-onset OTC deficiency: an underestimated mechanism of inheritance of an X-linked disease. Orphanet J Rare Dis 2024; 19:3. [PMID: 38167094 PMCID: PMC10763478 DOI: 10.1186/s13023-023-02997-8] [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: 05/10/2023] [Accepted: 12/13/2023] [Indexed: 01/05/2024] Open
Abstract
BACKGROUND Ornithine Transcarbamylase Deficiency (OTCD) is an X-linked urea cycle disorder characterized by acute hyperammonemic episodes. Hemizygous males are usually affected by a severe/fatal neonatal-onset form or, less frequently, by a late-onset form with milder disease course, depending on the residual enzymatic activity. Hyperammonemia can occur any time during life and patients could remain non- or mis-diagnosed due to unspecific symptoms. In heterozygous females, clinical presentation varies based on the extent of X chromosome inactivation. Maternal transmission in X-linked disease is the rule, but in late-onset OTCD, due to the milder phenotype of affected males, paternal transmission to the females is possible. So far, father-to-daughter transmission of OTCD has been reported only in 4 Japanese families. RESULTS We identified in 2 Caucasian families, paternal transmission of late-onset OTCD with severe/fatal outcome in affected males and 1 heterozygous female. Furthermore, we have reassessed the pedigrees of other published reports in 7 additional families with evidence of father-to-daughter inheritance of OTCD, identifying and listing the family members for which this transmission occurred. CONCLUSIONS Our study highlights how the diagnosis and pedigree analysis of late-onset OTCD may represent a real challenge for clinicians. Therefore, the occurrence of paternal transmission in OTCD should not be underestimated, due to the relevant implications for disease inheritance and risk of recurrence.
Collapse
Affiliation(s)
- Barbara Siri
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy.
- Department of Paediatrics, Città della Salute e della Scienza, OIRM, University of Turin, Turin, Italy.
| | - Giorgia Olivieri
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Francesca Romana Lepri
- Translational Cytogenomics Research Unit, Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Martin Poms
- Division of Clinical Chemistry and Biochemistry and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Bianca Maria Goffredo
- Division of Metabolism and Metabolic Diseases Research Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Anna Commone
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| | - Antonio Novelli
- Translational Cytogenomics Research Unit, Laboratory of Medical Genetics, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Johannes Häberle
- Division of Metabolism and Children's Research Center, University Children's Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Carlo Dionisi-Vici
- Division of Metabolic Diseases and Hepatology, Bambino Gesù Children's Hospital, IRCCS, Piazza S. Onofrio 4, 00165, Rome, Italy
| |
Collapse
|
37
|
Edelblut J, Skaar JR, Hilton J, Seibt M, Martin K, Hadker N, Quartel A, Steiner RD. Quantifying preferences for urea cycle disorder treatments using a discrete choice experiment. J Med Econ 2024; 27:506-517. [PMID: 38491962 DOI: 10.1080/13696998.2024.2330846] [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: 01/23/2024] [Accepted: 03/12/2024] [Indexed: 03/18/2024]
Abstract
AIMS Urea cycle disorders (UCDs) can cause ammonia accumulation and central nervous system toxicity. Nitrogen-binding medications can be efficacious, but certain attributes may negatively impact adherence. This study sought to quantify the administration-related attributes influencing overall prescription selection and patient adherence. METHODS A web-based, quantitative survey including discrete choice experiment (DCE) methodology captured responses from health care providers for patients with UCDs. A series of hypothetical treatment profile sets with attributes such as route of administration, taste/odor, preparation instructions, packaging, dose measurement, and weight use restrictions were presented. From 16 sets of 3 hypothetical product profiles, respondents evaluated attributes most preferred for prescription selection or patient adherence. Attributes assumed a higher overall preference if relative importance (RI) scores were >16.67% (the value if all attributes were of equal importance). Preference weight scores were assessed. A nine-point Likert scale assessed respondent attitudes, such as satisfaction. RESULTS A total of 51 respondents completed the survey. Respondents reported dissatisfaction with current treatments (mean [SD] = 5.4 [1.7]). For prescription selection, four attributes achieved RI >16.67%: taste/odor (24%), weight restrictions (21%), preparation instructions (18%), and route of administration (17%). For adherence, three attributes related to administration achieved RI >16.67%: taste/odor (28%), preparation instructions (21%), and route of administration (17%). Preference weights for "taste/odor masked" were higher than "not taste/odor masked" for prescription selection (mean [SD]; 1.52 [1.10] vs -1.52 [1.10]) and treatment adherence (73.8 [55.2] vs -73.8 [55.2]). LIMITATIONS This study contained a relatively small sample size. Survey respondent selection, the use of hypothetical product profiles, and exclusion of non-pharmacologic treatment options could have contributed to potential biases. CONCLUSIONS Among attributes tested, taste/odor was the most important attribute influencing overall preference for both prescribing and patient adherence, with taste/odor masking preferred. Optimizing nitrogen-binding medications through masking taste/odor may support improved patient adherence and outcomes in UCDs.
Collapse
Affiliation(s)
| | | | - John Hilton
- Acer Therapeutics, Newton, MA, USA, a wholly owned subsidiary of Zevra Therapeutics
| | - Matthew Seibt
- Acer Therapeutics, Newton, MA, USA, a wholly owned subsidiary of Zevra Therapeutics
| | | | | | - Adrian Quartel
- Acer Therapeutics, Newton, MA, USA, a wholly owned subsidiary of Zevra Therapeutics
| | - Robert D Steiner
- Department of Pediatrics, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| |
Collapse
|
38
|
Seidl MJ, Scharre S, Posset R, Druck AC, Epp F, Okun JG, Dimitrov B, Hoffmann GF, Kölker S, Zielonka M. ASS1 deficiency is associated with impaired neuronal differentiation in zebrafish larvae. Mol Genet Metab 2024; 141:108097. [PMID: 38113552 DOI: 10.1016/j.ymgme.2023.108097] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Revised: 11/22/2023] [Accepted: 11/23/2023] [Indexed: 12/21/2023]
Abstract
Citrullinemia type 1 (CTLN1) is a rare autosomal recessive urea cycle disorder caused by deficiency of the cytosolic enzyme argininosuccinate synthetase 1 (ASS1) due to pathogenic variants in the ASS1 gene located on chromosome 9q34.11. Even though hyperammenomia is considered the major pathomechanistic factor for neurological impairment and cognitive dysfunction, a relevant subset of individuals presents with a neurodegenerative course in the absence of hyperammonemic decompensations. Here we show, that ASS1 deficiency induced by antisense-mediated knockdown of the zebrafish ASS1 homologue is associated with defective neuronal differentiation ultimately causing neuronal cell loss and consecutively decreased brain size in zebrafish larvae in vivo. Whereas ASS1-deficient zebrafish larvae are characterized by markedly elevated concentrations of citrulline - the biochemical hallmark of CTLN1, accumulation of L-citrulline, hyperammonemia or therewith associated secondary metabolic alterations did not account for the observed phenotype. Intriguingly, coinjection of the human ASS1 mRNA not only normalized citrulline concentration but also reversed the morphological cerebral phenotype and restored brain size, confirming conserved functional properties of ASS1 across species. The results of the present study imply a novel, potentially non-enzymatic (moonlighting) function of the ASS1 protein in neurodevelopment.
Collapse
Affiliation(s)
- Marie J Seidl
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Svenja Scharre
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Roland Posset
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Ann-Catrin Druck
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Friederike Epp
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen G Okun
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Bianca Dimitrov
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Georg F Hoffmann
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Stefan Kölker
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Matthias Zielonka
- Heidelberg University, Medical Faculty Heidelberg, and Division of Pediatric Neurology and Metabolic Medicine, Center for Child and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany; Heidelberg Research Center for Molecular Medicine (HRCMM), Heidelberg, Germany.
| |
Collapse
|
39
|
Duff C, Alexander IE, Baruteau J. Gene therapy for urea cycle defects: An update from historical perspectives to future prospects. J Inherit Metab Dis 2024; 47:50-62. [PMID: 37026568 PMCID: PMC10953416 DOI: 10.1002/jimd.12609] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/08/2023]
Abstract
Urea cycle defects (UCDs) are severe inherited metabolic diseases with high unmet needs which present a permanent risk of hyperammonaemic decompensation and subsequent acute death or neurological sequelae, when treated with conventional dietetic and medical therapies. Liver transplantation is currently the only curative option, but has the potential to be supplanted by highly effective gene therapy interventions without the attendant need for life-long immunosuppression or limitations imposed by donor liver supply. Over the last three decades, pioneering genetic technologies have been explored to circumvent the consequences of UCDs, improve quality of life and long-term outcomes: adenoviral vectors, adeno-associated viral vectors, gene editing, genome integration and non-viral technology with messenger RNA. In this review, we present a summarised view of this historical path, which includes some seminal milestones of the gene therapy's epic. We provide an update about the state of the art of gene therapy technologies for UCDs and the current advantages and pitfalls driving future directions for research and development.
Collapse
Affiliation(s)
- Claire Duff
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
| | - Ian E. Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Faculty of Medicine and HealthThe University of Sydney and Sydney Children's Hospitals NetworkWestmeadNew South WalesAustralia
- Discipline of Child and Adolescent HealthThe University of SydneyWestmeadNew South WalesAustralia
| | - Julien Baruteau
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child HealthUniversity College LondonLondonUK
- National Institute of Health Research Great Ormond Street Biomedical Research CentreLondonUK
- Metabolic Medicine DepartmentGreat Ormond Street Hospital for Children NHS Foundation TrustLondonUK
| |
Collapse
|
40
|
Olofsson S, Löfvendahl S, Widén J, Rudebeck M, Lindgren P, Stepien KM, Arnoux JB, Luz Couce Pico M, Leão Teles E, Jacobson L. Societal costs and quality of life associated with arginase 1 deficiency in a European setting - a multinational, cross-sectional survey. J Med Econ 2024; 27:1146-1156. [PMID: 39230682 DOI: 10.1080/13696998.2024.2400856] [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: 07/09/2024] [Revised: 08/29/2024] [Accepted: 09/02/2024] [Indexed: 09/05/2024]
Abstract
BACKGROUND AND AIMS Arginase 1 deficiency (ARG1-D) is a ultrarare disease with manifestations that cause mobility and cognitive impairment that progress over time and may lead to early mortality. Diseases such as ARG1-D have a major impact also outside of the health care sector and the aim of this study was to estimate the current burden of disease associated with ARG1-D from a societal perspective. METHODS The study was performed as a web-based survey of patients with ARG1-D and their caregivers in four European countries (France, Portugal, Spain, United Kingdom). The survey was distributed at participating clinics and included questions on e.g. symptoms (including the Gross Motor Function Classification System, GMFCS, and cognitive impairment), health care use, medication, ability to work, caregiving, and impact on health-related quality-of-life (HRQoL) using the EQ-5D-5L. RESULTS The estimated total mean societal cost per patient and year was £63,775 (SD: £49,944). The cost varied significantly with both mobility impairment (from £49,809 for GMFCS level 1 to £103,639 for GMFCS levels 3-5) and cognitive impairment (from £43,860 for mild level to £99,162 for severe level). The mean utility score on the EQ-5D-5L for patients was 0.498 (SD: 0.352). The utility score also varied significantly with both mobility impairment (from 0.783 for GMFCS level 1 to 0.153 for GMFCS level 3-5) and cognitive impairment (from 0.738 for mild level to 0.364 for severe level). CONCLUSIONS Similar to other studies of rare diseases, the study is based on a limited number of observations. However, the sample appear to be reasonably representative when comparing to previous studies of ARG1-D. This study shows that ARG1-D is associated with a high societal cost and significant impact on HRQoL. Earlier diagnosis and better treatment options that can postpone or withhold progression may therefore have a potential for improved HRQoL and savings for the patient, caregiver, and society.
Collapse
Affiliation(s)
- Sara Olofsson
- The Swedish Institute for Health Economics, IHE, Lund, Sweden
| | | | | | | | - Peter Lindgren
- The Swedish Institute for Health Economics, IHE, Lund, Sweden
- Karolinska Institutet, Stockholm, Sweden
| | - Karolina M Stepien
- Salford Royal Organization, Northern Care Alliance NHS Foundation Trust, Salford, United Kingdom
| | | | - Maria Luz Couce Pico
- Hospital Clinico Universitario de Santiago de Compostela, IDIS, MetabERN, Santiago de Compostela, Spain
| | - Elisa Leão Teles
- Centro Hospitalar Universitário de São João, MetabERN, Porto, Portugal
| | | |
Collapse
|
41
|
Seker Yilmaz B, Baruteau J, Chakrapani A, Champion M, Chronopoulou E, Claridge LC, Daly A, Davies C, Davison J, Dhawan A, Grunewald S, Gupte GL, Heaton N, Lemonde H, McKiernan P, Mills P, Morris AA, Mundy H, Pierre G, Rajwal S, Sivananthan S, Sreekantam S, Stepien KM, Vara R, Yeo M, Gissen P. Liver transplantation in ornithine transcarbamylase deficiency: A retrospective multicentre cohort study. Mol Genet Metab Rep 2023; 37:101020. [PMID: 38053940 PMCID: PMC10694733 DOI: 10.1016/j.ymgmr.2023.101020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
Ornithine transcarbamylase deficiency (OTCD) is an X-linked defect of ureagenesis and the most common urea cycle disorder. Patients present with hyperammonemia causing neurological symptoms, which can lead to coma and death. Liver transplantation (LT) is the only curative therapy, but has several limitations including organ shortage, significant morbidity and requirement of lifelong immunosuppression. This study aims to identify the characteristics and outcomes of patients who underwent LT for OTCD. We conducted a retrospective study for OTCD patients from 5 UK centres receiving LT in 3 transplantation centres between 2010 and 2022. Patients' demographics, family history, initial presentation, age at LT, graft type and pre- and post-LT clinical, metabolic, and neurocognitive profile were collected from medical records. A total of 20 OTCD patients (11 males, 9 females) were enrolled in this study. 6/20 had neonatal and 14/20 late-onset presentation. 2/20 patients had positive family history for OTCD and one of them was diagnosed antenatally and received prospective treatment. All patients were managed with standard of care based on protein-restricted diet, ammonia scavengers and supplementation with arginine and/or citrulline before LT. 15/20 patients had neurodevelopmental problems before LT. The indication for LT was presence (or family history) of recurrent metabolic decompensations occurring despite standard medical therapy leading to neurodisability and quality of life impairment. Median age at LT was 10.5 months (6-24) and 66 months (35-156) in neonatal and late onset patients, respectively. 15/20 patients had deceased donor LT (DDLT) and 5/20 had living related donor LT (LDLT). Overall survival was 95% with one patient dying 6 h after LT. 13/20 had complications after LT and 2/20 patients required re-transplantation. All patients discontinued dietary restriction and ammonia scavengers after LT and remained metabolically stable. Patients who had neurodevelopmental problems before LT persisted to have difficulties after LT. 1/5 patients who was reported to have normal neurodevelopment before LT developed behavioural problems after LT, while the remaining 4 maintained their abilities without any reported issues. LT was found to be effective in correcting the metabolic defect, eliminates the risk of hyperammonemia and prolongs patients' survival.
Collapse
Affiliation(s)
- Berna Seker Yilmaz
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Julien Baruteau
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Department of Paediatric Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Anupam Chakrapani
- Department of Paediatric Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Michael Champion
- Department of Inherited Metabolic Disease, Evelina Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, SE1 7EH London, UK
| | - Efstathia Chronopoulou
- Department of Inherited Metabolic Disease, Division of Women's and Children's Services, University Hospitals Bristol NHS Foundation Trust, Bristol BS1 3NU, UK
| | | | - Anne Daly
- Birmingham Women's and Children's Hospital NHS Foundation Trust, B4 6NH, Birmingham, UK
| | - Catherine Davies
- Department of Inherited Metabolic Disease, Evelina Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, SE1 7EH London, UK
| | - James Davison
- Department of Paediatric Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Anil Dhawan
- Paediatric Liver Gastroenterology and Nutrition Centre and Mowat Labs, King's College Hospital NHS Foundation Trust, WC2R 2LS, London, UK
| | - Stephanie Grunewald
- Department of Paediatric Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Girish L. Gupte
- Birmingham Women's and Children's Hospital NHS Foundation Trust, B4 6NH, Birmingham, UK
| | - Nigel Heaton
- Institute of Liver Studies, Kings College Hospital, Denmark Hill, WC2R 2LS London, UK
| | - Hugh Lemonde
- Department of Inherited Metabolic Disease, Evelina Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, SE1 7EH London, UK
| | - Pat McKiernan
- Birmingham Women's and Children's Hospital NHS Foundation Trust, B4 6NH, Birmingham, UK
| | - Philippa Mills
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
| | - Andrew A.M. Morris
- Willink Unit, Genetic Medicine, Manchester Academic Health Sciences Centre, Central Manchester University Hospitals NHS Foundation Trust, Oxford Road, Manchester M13 9WL, UK
| | - Helen Mundy
- Department of Inherited Metabolic Disease, Evelina Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, SE1 7EH London, UK
| | - Germaine Pierre
- Department of Inherited Metabolic Disease, Division of Women's and Children's Services, University Hospitals Bristol NHS Foundation Trust, Bristol BS1 3NU, UK
| | - Sanjay Rajwal
- Leeds Teaching Hospitals NHS Trust, LS9 7TF Leeds, UK
| | - Siyamini Sivananthan
- Department of Paediatric Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Srividya Sreekantam
- Birmingham Women's and Children's Hospital NHS Foundation Trust, B4 6NH, Birmingham, UK
| | - Karolina M. Stepien
- Adult Inherited Metabolic Diseases, Salford Royal NHS Foundation Trust, M6 8HD Salford, UK
| | - Roshni Vara
- Department of Inherited Metabolic Disease, Evelina Children's Hospital, Guy's and St Thomas' NHS Foundation Trust, SE1 7EH London, UK
| | - Mildrid Yeo
- Department of Paediatric Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| | - Paul Gissen
- Genetics and Genomic Medicine Department, Great Ormond Street Institute of Child Health, University College London, London WC1N 1EH, UK
- Department of Paediatric Metabolic Medicine, Great Ormond Street Hospital for Children NHS Foundation Trust, London WC1N 3JH, UK
| |
Collapse
|
42
|
Zhang Z, Tong F, Chen C, Zhang T, Qian G, Yang X, Huang X, Yang R, Zhao Z. Long-term follow-up of children with carbamoyl phosphate synthase 1 deficiency detected in newborn screening. Zhejiang Da Xue Xue Bao Yi Xue Ban 2023; 52:721-726. [PMID: 37986659 PMCID: PMC10764181 DOI: 10.3724/zdxbyxb-2023-0359] [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: 08/01/2023] [Accepted: 10/05/2023] [Indexed: 11/22/2023]
Abstract
OBJECTIVES To investigate genotype-phenotype characteristics and long-term prognosis of neonatal carbamoyl phosphate synthetase 1 (CPS1) deficiency among children through newborn screening in Zhejiang province. METHODS The clinical and follow-up data of children with CPS1 deficiency detected through neonatal screening and confirmed by tandem mass spectrometry and genetic testing in Zhejiang Province Newborn Disease Screening Center from September 2013 to August 2023 were retrospectively analyzed. RESULTS A total of 4 056 755 newborns were screened and 6 cases of CPS1 deficiency were diagnosed through phenotypic and genetic testing. Ten different variations of CPS1 genewere identified in genetic testing, including 2 known pathogenic variations (c.2359C>T and c.1549+1G>T) and 8 unreported variations (c.3405-1G>T, c.2372C>T, c.1436C>T, c.2228T>C, c.2441G>A, c.3031G>A, c.3075T>C and c.390-403del). All patients had decreased citrulline levels (2.72-6.21 μmol/L), and varying degrees of elevated blood ammonia. The patients received restricted natural protein intake (special formula), arginine and supportive therapy after diagnosis, and were followed-up for a period ranging from 9 months to 10 years. Three patients experienced hyperammonemia, and one patient each had attention deficit hyperactivity disorder, transient facial twitching and increased muscle tone. One patient died, while the other five surviving patients had normal scores of the Ages & Stages Questionnaires (ASQ) and Griffiths Development Scales up to the present time; 4 cases had combined height or weight lag and one case was normal in height and weight. CONCLUSIONS Low citrulline levels and hyperammonemia are common in CPS1 deficiency patients in Zhejiang. Most gene variants identified were specific to individual families, and no hotspot mutations were found. Early diagnosis through newborn screening and following standardized treatment can significantly improve the prognosis of the patients.
Collapse
Affiliation(s)
- Zhanming Zhang
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China.
| | - Fan Tong
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Chi Chen
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Ting Zhang
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Guling Qian
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Xin Yang
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Xinwen Huang
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Rulai Yang
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China
| | - Zhengyan Zhao
- Department of Genetic and Metabolism,Children's Hospital,Zhejiang University School of Medicine,National Clinical Research Center for Child Health, Hangzhou 310052, China.
| |
Collapse
|
43
|
Murali CN, Barber JR, McCarter R, Zhang A, Gallant N, Simpson K, Dorrani N, Wilkening GN, Hays RD, Lichter-Konecki U, Burrage LC, Nagamani SCS. Health-related quality of life in a systematically assessed cohort of children and adults with urea cycle disorders. Mol Genet Metab 2023; 140:107696. [PMID: 37690181 PMCID: PMC10866211 DOI: 10.1016/j.ymgme.2023.107696] [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: 06/21/2023] [Revised: 09/01/2023] [Accepted: 09/02/2023] [Indexed: 09/12/2023]
Abstract
PURPOSE Individuals with urea cycle disorders (UCDs) may develop recurrent hyperammonemia, episodic encephalopathy, and neurological sequelae which can impact Health-related Quality of Life (HRQoL). To date, there have been no systematic studies of HRQoL in people with UCDs. METHODS We reviewed HRQoL and clinical data for 190 children and 203 adults enrolled in a multicenter UCD natural history study. Physical and psychosocial HRQoL in people with UCDs were compared to HRQoL in healthy people and people with phenylketonuria (PKU) and diabetes mellitus. We assessed relationships between HRQoL, UCD diagnosis, and disease severity. Finally, we calculated sample sizes required to detect changes in these HRQoL measures. RESULTS Individuals with UCDs demonstrated worse physical and psychosocial HRQoL than their healthy peers and peers with PKU and diabetes. In children, HRQoL scores did not differ by diagnosis or severity. In adults, individuals with decreased severity had worse psychosocial HRQoL. Finally, we show that a large number of individuals would be required in clinical trials to detect differences in HRQoL in UCDs. CONCLUSION Individuals with UCDs have worse HRQoL compared to healthy individuals and those with PKU and diabetes. Future work should focus on the impact of liver transplantation and other clinical variables on HRQoL in UCDs.
Collapse
Affiliation(s)
- Chaya N Murali
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - John R Barber
- Children's National Health System, Washington, DC, USA
| | | | - Anqing Zhang
- Children's National Health System, Washington, DC, USA
| | - Natalie Gallant
- Department of Pediatrics, University of California Irvine, Irvine, CA, USA
| | - Kara Simpson
- Children's National Health System, Washington, DC, USA
| | - Naghmeh Dorrani
- Department of Human Genetics, University of California Los Angeles, Los Angeles, CA, USA
| | | | - Ron D Hays
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Uta Lichter-Konecki
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Lindsay C Burrage
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA
| | - Sandesh C S Nagamani
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
44
|
Abou Haidar L, Pachnis P, Gotway GK, Ni M, DeBerardinis RJ, McNutt MC. Partial N-acetyl glutamate synthase deficiency presenting as postpartum hyperammonemia: Diagnosis and subsequent pregnancy management. JIMD Rep 2023; 64:403-409. [PMID: 37927481 PMCID: PMC10623101 DOI: 10.1002/jmd2.12388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Revised: 07/28/2023] [Accepted: 08/01/2023] [Indexed: 11/07/2023] Open
Abstract
N-acetyl glutamate synthase (NAGS) deficiency (OMIM #: 237310) is a rare urea cycle disorder that usually presents early in life with hyperammonemia. NAGS catalyzes the synthesis of N-acetyl glutamate (NAG) which functions as an activator of the carbamoyl phosphate synthetase-1 mediated conversion of ammonia to carbamoyl phosphate. The absence of NAG results in a proximal urea cycle disorder which can result in severe neurologic sequelae secondary to hyperammonemia and even death. Unlike the other urea cycle disorders, a specific pharmacological treatment for NAGS deficiency exists in the form of carglumic acid, an analog of NAG. Here we present a 29-year-old previously healthy female who presented with hyperammonemia and obtundation just after the birth of her first child. Exome sequencing revealed two novel variants in the NAGS gene, and plasma metabolomics revealed extremely low levels of NAG. Carglumic acid treatment led to prompt resolution of her biochemical abnormalities and symptoms. She tolerated two subsequent pregnancies, 2 years and 6 years after her initial presentation, while taking carglumic acid, and breastfed her third child, all without complications in the mother or children. This case report emphasizes the importance of considering urea cycle disorders in previously-healthy adults presenting with neurological symptoms during periods of metabolic stress, including the postpartum period. It also highlights the efficacious and safe use of carglumic acid during pregnancy and while breastfeeding.
Collapse
Affiliation(s)
- Lea Abou Haidar
- Children's Medical Center Research InstituteThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Howard Hughes Medical InstituteThe University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Panayotis Pachnis
- Children's Medical Center Research InstituteThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Department of PediatricsThe University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Garrett K. Gotway
- Department of PediatricsThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Eugene McDermott Center for Human Growth and DevelopmentThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Department of Internal MedicineThe University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Min Ni
- Children's Medical Center Research InstituteThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Department of PediatricsThe University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Ralph J. DeBerardinis
- Children's Medical Center Research InstituteThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Howard Hughes Medical InstituteThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Department of PediatricsThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Eugene McDermott Center for Human Growth and DevelopmentThe University of Texas Southwestern Medical CenterDallasTexasUSA
| | - Markey C. McNutt
- Department of PediatricsThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Eugene McDermott Center for Human Growth and DevelopmentThe University of Texas Southwestern Medical CenterDallasTexasUSA
- Department of Internal MedicineThe University of Texas Southwestern Medical CenterDallasTexasUSA
| |
Collapse
|
45
|
Loeffler J, Elfiky A, Al Moussawi H, Ravindran N. Hyperammonemia Encephalopathy due to Urea Cycle Disorder Precipitated by Gastrointestinal Bleed in the Setting of Prior Bariatric Surgery. ACG Case Rep J 2023; 10:e01164. [PMID: 37799484 PMCID: PMC10550022 DOI: 10.14309/crj.0000000000001164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 08/29/2023] [Indexed: 10/07/2023] Open
Abstract
The urea cycle is a metabolic pathway that excretes nitrogenous waste products from the body. Urea cycle disorders (UCDs) result from enzymatic deficiencies within this pathway, which can lead to life-threatening hyperammonemia. Gastric bypass-related hyperammonemia in patients who have undergone Roux-en-Y gastric bypass surgery has been previously reported. UCDs have been implicated as a cause of gastric bypass-related hyperammonemia. In this report, we present the case of a patient with a history of bariatric surgery who experienced severe hyperammonemia encephalopathy triggered by a gastrointestinal bleed due to an undiagnosed UCD.
Collapse
Affiliation(s)
| | - Ahmed Elfiky
- Department of Gastroenterology and Hepatology, Staten Island University Hospital, NY
| | - Hassan Al Moussawi
- Department of Gastroenterology and Hepatology, Staten Island University Hospital, NY
| | - Nishal Ravindran
- Department of Gastroenterology and Hepatology, Staten Island University Hospital, NY
| |
Collapse
|
46
|
Kho J, Polak U, Jiang MM, Odom JD, Hunter JV, Ali SM, Burrage LC, Nagamani SC, Pautler RG, Thompson HP, Urayama A, Jin Z, Lee B. Argininosuccinate lyase deficiency causes blood-brain barrier disruption via nitric oxide-mediated dysregulation of claudin expression. JCI Insight 2023; 8:e168475. [PMID: 37490345 PMCID: PMC10544197 DOI: 10.1172/jci.insight.168475] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023] Open
Abstract
Nitric oxide (NO) is a critical signaling molecule that has been implicated in the pathogenesis of neurocognitive diseases. Both excessive and insufficient NO production have been linked to pathology. Previously, we have shown that argininosuccinate lyase deficiency (ASLD) is a novel model system to investigate cell-autonomous, nitric oxide synthase-dependent NO deficiency. Humans with ASLD are at increased risk for developing hyperammonemia due to a block in ureagenesis. However, natural history studies have shown that individuals with ASLD have multisystem disease including neurocognitive deficits that can be independent of ammonia. Here, using ASLD as a model of NO deficiency, we investigated the effects of NO on brain endothelial cells in vitro and the blood-brain barrier (BBB) in vivo. Knockdown of ASL in human brain microvascular endothelial cells (HBMECs) led to decreased transendothelial electrical resistance, indicative of increased cell permeability. Mechanistically, treatment with an NO donor or inhibition of Claudin-1 improved barrier integrity in ASL-deficient HBMECs. Furthermore, in vivo assessment of a hypomorphic mouse model of ASLD showed increased BBB leakage, which was partially rescued by NO supplementation. Our results suggest that ASL-mediated NO synthesis is required for proper maintenance of brain microvascular endothelial cell functions as well as BBB integrity.
Collapse
Affiliation(s)
- Jordan Kho
- Department of Molecular and Human Genetics and
| | | | | | | | - Jill V. Hunter
- Department of Radiology, Baylor College of Medicine, Houston, Texas, USA
- Texas Children’s Hospital, Houston, Texas, USA
| | | | - Lindsay C. Burrage
- Department of Molecular and Human Genetics and
- Texas Children’s Hospital, Houston, Texas, USA
| | - Sandesh C.S. Nagamani
- Department of Molecular and Human Genetics and
- Texas Children’s Hospital, Houston, Texas, USA
| | - Robia G. Pautler
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, Texas, USA
| | - Hannah P. Thompson
- Department of Neurology, University of Texas Health Science Center, Houston, Texas, USA
| | - Akihiko Urayama
- Department of Neurology, University of Texas Health Science Center, Houston, Texas, USA
| | - Zixue Jin
- Department of Molecular and Human Genetics and
| | - Brendan Lee
- Department of Molecular and Human Genetics and
- Texas Children’s Hospital, Houston, Texas, USA
| |
Collapse
|
47
|
Ladha FA, Le Mons C, Craigen WJ, Magoulas PL, Marom R, Lewis AM. Barriers to a successful healthcare transition for individuals with urea cycle disorders. Mol Genet Metab 2023; 139:107609. [PMID: 37245377 PMCID: PMC11955086 DOI: 10.1016/j.ymgme.2023.107609] [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: 04/03/2023] [Revised: 05/13/2023] [Accepted: 05/14/2023] [Indexed: 05/30/2023]
Abstract
The pediatric to adult healthcare transition (HCT) is a process for individuals with chronic health conditions to gradually shift from a pediatric to an adult-oriented care system. Autonomy and self-management skills required for an individual's HCT readiness can be evaluated through the transition readiness assessment questionnaire (TRAQ). Despite general HCT preparation guidelines, little is known about the HCT experience of individuals with a urea cycle disorder (UCD). This is the first study to report the parent or guardian perception of the HCT process in children with a UCD by investigating the stages of transition readiness and transition outcome. We identify barriers to HCT readiness and planning, along with deficiencies in transition outcome for individuals with a UCD. For children that received special education services compared to those that did not, significantly lower transition readiness scores were identified in the total TRAQ score (p = 0.03) and in the domains of tracking health issues (p = 0.02), talking with providers (p = 0.03), and managing daily activities (p = 0.01). There was a lack of HCT preparation as most subjects did not have a HCT discussion with their healthcare provider before age 26. Deficiencies in HCT outcome are demonstrated by individuals with a UCD reporting delays in needed medical care and dissatisfaction with their healthcare services. Considerations for facilitating a successful HCT for individuals with a UCD include providing individualized education, appointing a transition coordinator, allowing flexibility in HCT timing, and ensuring that the individual recognizes concerning UCD symptoms and knows when to seek medical care.
Collapse
Affiliation(s)
- Farah A Ladha
- School of Health Professions, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA.
| | - Cynthia Le Mons
- National Urea Cycle Disorders Foundation, Pasadena, CA, USA.
| | - William J Craigen
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA.
| | - Pilar L Magoulas
- School of Health Professions, Baylor College of Medicine, Houston, TX, USA; Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA.
| | - Ronit Marom
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA.
| | - Andrea M Lewis
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA; Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
48
|
Daly O, Mahiny AJ, Majeski S, McClintock K, Reichert J, Boros G, Szabó GT, Reinholz J, Schreiner P, Reid S, Lam K, Lepper M, Adler M, Meffen T, Heyes J, Karikó K, Lutwyche P, Vlatkovic I. ASL mRNA-LNP Therapeutic for the Treatment of Argininosuccinic Aciduria Enables Survival Benefit in a Mouse Model. Biomedicines 2023; 11:1735. [PMID: 37371829 DOI: 10.3390/biomedicines11061735] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Revised: 06/05/2023] [Accepted: 06/11/2023] [Indexed: 06/29/2023] Open
Abstract
Argininosuccinic aciduria (ASA) is a metabolic disorder caused by a deficiency in argininosuccinate lyase (ASL), which cleaves argininosuccinic acid to arginine and fumarate in the urea cycle. ASL deficiency (ASLD) leads to hepatocyte dysfunction, hyperammonemia, encephalopathy, and respiratory alkalosis. Here we describe a novel therapeutic approach for treating ASA, based on nucleoside-modified messenger RNA (modRNA) formulated in lipid nanoparticles (LNP). To optimize ASL-encoding mRNA, we modified its cap, 5' and 3' untranslated regions, coding sequence, and the poly(A) tail. We tested multiple optimizations of the formulated mRNA in human cells and wild-type C57BL/6 mice. The ASL protein showed robust expression in vitro and in vivo and a favorable safety profile, with low cytokine and chemokine secretion even upon administration of increasing doses of ASL mRNA-LNP. In the ASLNeo/Neo mouse model of ASLD, intravenous administration of the lead therapeutic candidate LNP-ASL CDS2 drastically improved the survival of the mice. When administered twice a week lower doses partially protected and 3 mg/kg LNP-ASL CDS2 fully protected the mice. These results demonstrate the considerable potential of LNP-formulated, modified ASL-encoding mRNA as an effective alternative to AAV-based approaches for the treatment of ASA.
Collapse
Affiliation(s)
- Owen Daly
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | | | - Sara Majeski
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | | | | | - Gábor Boros
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| | | | | | - Petra Schreiner
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Steve Reid
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Kieu Lam
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - Marlen Lepper
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| | - Melanie Adler
- BioNTech SE, An der Goldgrube 12, 55131 Mainz, Germany
| | - Tracy Meffen
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | - James Heyes
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | | | - Pete Lutwyche
- Genevant Sciences Corporation, Vancouver, BC V5T 4T5, Canada
| | | |
Collapse
|
49
|
Lo RS, Cromie GA, Tang M, Teng K, Owens K, Sirr A, Kutz JN, Morizono H, Caldovic L, Ah Mew N, Gropman A, Dudley AM. The functional impact of 1,570 individual amino acid substitutions in human OTC. Am J Hum Genet 2023; 110:863-879. [PMID: 37146589 PMCID: PMC10183466 DOI: 10.1016/j.ajhg.2023.03.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Accepted: 03/30/2023] [Indexed: 05/07/2023] Open
Abstract
Deleterious mutations in the X-linked gene encoding ornithine transcarbamylase (OTC) cause the most common urea cycle disorder, OTC deficiency. This rare but highly actionable disease can present with severe neonatal onset in males or with later onset in either sex. Individuals with neonatal onset appear normal at birth but rapidly develop hyperammonemia, which can progress to cerebral edema, coma, and death, outcomes ameliorated by rapid diagnosis and treatment. Here, we develop a high-throughput functional assay for human OTC and individually measure the impact of 1,570 variants, 84% of all SNV-accessible missense mutations. Comparison to existing clinical significance calls, demonstrated that our assay distinguishes known benign from pathogenic variants and variants with neonatal onset from late-onset disease presentation. This functional stratification allowed us to identify score ranges corresponding to clinically relevant levels of impairment of OTC activity. Examining the results of our assay in the context of protein structure further allowed us to identify a 13 amino acid domain, the SMG loop, whose function appears to be required in human cells but not in yeast. Finally, inclusion of our data as PS3 evidence under the current ACMG guidelines, in a pilot reclassification of 34 variants with complete loss of activity, would change the classification of 22 from variants of unknown significance to clinically actionable likely pathogenic variants. These results illustrate how large-scale functional assays are especially powerful when applied to rare genetic diseases.
Collapse
Affiliation(s)
- Russell S Lo
- Pacific Northwest Research Institute, Seattle, WA, USA
| | | | - Michelle Tang
- Pacific Northwest Research Institute, Seattle, WA, USA
| | - Kevin Teng
- Pacific Northwest Research Institute, Seattle, WA, USA
| | - Katherine Owens
- Pacific Northwest Research Institute, Seattle, WA, USA; Department of Applied Mathematics, University of Washington, Seattle, WA, USA
| | - Amy Sirr
- Pacific Northwest Research Institute, Seattle, WA, USA
| | - J Nathan Kutz
- Department of Applied Mathematics, University of Washington, Seattle, WA, USA
| | - Hiroki Morizono
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, USA; Department of Genomics and Precision Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Ljubica Caldovic
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, USA; Department of Genomics and Precision Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Nicholas Ah Mew
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, USA; Department of Genomics and Precision Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA
| | - Andrea Gropman
- Center for Genetic Medicine Research, Children's National Research Institute, Children's National Hospital, Washington, DC, USA; Department of Genomics and Precision Medicine, School of Medicine and Health Sciences, The George Washington University, Washington, DC, USA; Department of Neurology, Division of Neurogenetics and Neurodevelopmental Disabilities, Children's National Hospital, Washington, DC, USA; Center for Neuroscience Research, Children's National Research Institute, Children's National Hospital, Washington, DC, USA
| | | |
Collapse
|
50
|
Forsyth R, Peretz RH, Dempsey A, Britton J, Kratz L, Hamosh A, Vernon H, Batshaw ML, Valle D. The remarkable journey of one female individual with ornithine transcarbamylase deficiency diagnosed post-mortem. JIMD Rep 2023; 64:233-237. [PMID: 37151362 PMCID: PMC10159862 DOI: 10.1002/jmd2.12361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/08/2022] [Revised: 01/12/2023] [Accepted: 01/16/2023] [Indexed: 01/31/2023] Open
Abstract
Urea cycle disorders (UCDs) comprise a group of inborn errors of metabolism with impaired ammonia clearance and an incidence of ~1:35 000 individuals. First described in the 1970s, the diagnosis and management of these disorders has evolved dramatically. We report on a 59-year-old woman with a UCD who contributed to advances in the understanding and treatment of this group of disorders. This individual was diagnosed with carbamoyl phosphate synthetase 1 deficiency based on a biochemical assay under a research context predating genetic sequencing, treated longitudinally as having this metabolic disorder, and was among the first participants to trial UCD pharmaceutical therapies. She ultimately succumbed to a SARS-CoV-2 infection while maintaining unexpectedly normal ammonium levels. Postmortem genetic testing revealed ornithine transcarbamylase deficiency. This individual's contributions to the field of UCDs is discussed herein.
Collapse
Affiliation(s)
- RaeLynn Forsyth
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Ryan H. Peretz
- National Human Genome Research InstituteNational Institutes of HealthBethesdaMarylandUSA
| | - Angela Dempsey
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Jacquelyn Britton
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Lisa Kratz
- Biochemical Genetics LaboratoryKennedy Krieger InstituteBaltimoreMarylandUSA
| | - Ada Hamosh
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Hilary Vernon
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Mark L. Batshaw
- Center for Genetic Medicine ResearchChildren's National HospitalWashingtonDCUSA
| | - David Valle
- Department of Genetic MedicineJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|