1
|
Takanashi JI. Magnetic resonance imaging and spectroscopy in hypomyelinating leukodystrophy. Brain Dev 2025; 47:104345. [PMID: 40174481 DOI: 10.1016/j.braindev.2025.104345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/03/2025] [Accepted: 03/03/2025] [Indexed: 04/04/2025]
Abstract
Recent advancements in molecular biology and radiology have led to the identification of several new leukodystrophies. A key diagnostic feature of leukodystrophies is the increased white matter signal intensity observed on T2-weighted magnetic resonance (MR) images. Leukodystrophies are typically classified into two main categories: hypomyelinating leukodystrophies (HLD) and other forms, including demyelinating leukodystrophies. HLD is characterized by a primary defect in myelin due to genetic variants that affect structural myelin proteins, oligodendrocyte transcription factors, RNA translation, and lysosomal proteins. Radiologically, HLD tends to show less pronounced white matter hyperintensity on T2-weighted images than demyelinating leukodystrophies. A definitive diagnosis can often be made by identifying abnormalities in regions beyond the white matter, such as the basal ganglia or cerebellum, or through the presence of characteristic clinical symptoms. N-acetylaspartate, a neuroaxonal marker observed on MR spectroscopy, is typically reduced in many neurological conditions, but N-acetylaspartate levels often remain normal in HLD, which is considered a distinctive feature of this disorder. This article provides an overview of the latest imaging findings and clinical features associated with HLD.
Collapse
Affiliation(s)
- Jun-Ichi Takanashi
- Department of Pediatrics, Tokyo Women's Medical University Yachiyo Medical Center, 477-96 Owadashinden, Yachiyo-Shi, Chiba 276-8524, Japan.
| |
Collapse
|
2
|
Kaufman C, D'Andrea A, Hackenberg A, Poms M, Braissant O, Häberle J. Diagnostic delay in cerebral creatine deficiency disorders: lessons learned from a cross-sectional single center study, and guanidinoacetate and creatine measurements in Switzerland between 2015 and 2023. Mol Cell Pediatr 2025; 12:1. [PMID: 39838169 PMCID: PMC11751272 DOI: 10.1186/s40348-024-00188-4] [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: 10/24/2024] [Accepted: 12/13/2024] [Indexed: 01/23/2025] Open
Abstract
BACKGROUND Cerebral creatine deficiency disorders (CCDD) are rare diseases caused by defects in the enzymes L-arginine: glycine amidinotransferase (AGAT) or guanidinoacetate-N-methyltransferase (GAMT), which are involved in synthesis of creatine; or by a defect in the creatine transporter (CRTR), which is essential for uptake of creatine as important energy source into the target cells. Patients with CCDD can present with a variety of unspecific symptoms: global developmental delay, speech-language disorder, behavioral abnormalities and seizures. Early treatment initiation is essential in AGAT and GAMT deficiencies to achieve a favorable outcome. This study describes the CCDD patient cohort in a single center, and an analysis of the referrals to two Swiss laboratories in Lausanne and Zurich between 2015 and 2023 for the two marker metabolites guanidinoacetate and creatine. RESULTS The patient cohort comprised 6 patients (defects: 2 GAMT, 4 CRTR), who were initially seen by different subspecialties depending on first symptoms. There was a diagnostic and therapeutic delay between 3 and 32 months (mean 13.8). Numbers of biomarker requests showed a constant increase during the study period, with a majority of tests performed in urine, the preferred sample for CCDD detection. Almost all samples (93.3%) were sent in by large hospitals (mainly from neurology, developmental pediatrics and metabolism) and only few (5.2%) by pediatricians in private practice, although those usually see the patients first. CONCLUSIONS The data from this study demonstrate a relevant delay in identifying patients with these rare conditions, and a predominance of biomarker analysis requested from pediatric subspecialties that are involved in patient management often long after occurrence of symptoms. To reduce the diagnostic delay and the outcome of patients, the current practice of sample referral should be reflected and first-contact healthcare providers should be encouraged to initiate selective screening.
Collapse
Affiliation(s)
- Christina Kaufman
- Division of Metabolism and Children`s Research Center, University Children's Hospital Zurich, Lenggstr. 30, 8008, Zurich, Switzerland
| | - Anaïs D'Andrea
- Division of Metabolism and Children`s Research Center, University Children's Hospital Zurich, Lenggstr. 30, 8008, Zurich, Switzerland
| | - Annette Hackenberg
- Division of Pediatric Neurology, University Children's Hospital Zurich, Zurich, Switzerland
| | - Martin Poms
- Division of Clinical Chemistry and Biochemistry, University Children's Hospital Zurich, Zurich, Switzerland
| | - Olivier Braissant
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Johannes Häberle
- Division of Metabolism and Children`s Research Center, University Children's Hospital Zurich, Lenggstr. 30, 8008, Zurich, Switzerland.
| |
Collapse
|
3
|
Tkachyova I, Tropak MB, Lee A, Datti A, Ito S, Schulze A. Targeting AGAT gene expression - a drug screening approach for the treatment of GAMT deficiency. Expert Opin Drug Discov 2024; 19:1383-1397. [PMID: 39402976 DOI: 10.1080/17460441.2024.2412994] [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: 06/21/2024] [Accepted: 10/02/2024] [Indexed: 11/13/2024]
Abstract
BACKGROUND Targeting the enzyme L-Arginine:glycine amidinotransferase (AGAT) to reduce the formation of guanidinoacetate (GAA) in patients with guanidinoacetate methyltransferase (GAMT) deficiency, we attempted to identify drugs for repurposing that reduce the expression of AGAT via transcriptional inhibition. RESEARCH DESIGN AND METHODS The authors applied a HeLa cell line stably expressing AGAT promoter and firefly luciferase reporter for high-content screening and secondary screening. For further assessment, the authors integrated Nanoluc luciferase as a reporter into the endogenous AGAT gene in HAP1 cell lines and used the human immortalized cell line RH30 as model of GAMT deficiency. RESULTS Screening 6,000 drugs and drug-like compounds, the authors identified 43 and 34 high-score candidates as inhibitors and inducers of AGAT promoter-reporter expression, respectively. After further deselection considering dose response, drug toxicity, topical formulations, price, and accessibility, the authors assessed seven candidates and found none of them demonstrating efficacy in HAP1 and RH30 cells and warranting further assessment. CONCLUSION The selection of the test models is crucial for screening of gene repressor drugs. Almost all drugs with an impact on gene expression had off-target effects. It is unlikely to find drugs that are selective inhibitors of AGAT expression, rendering pharmacological AGAT gene repression a risky approach for the treatment of GAMT deficiency.
Collapse
Affiliation(s)
- Ilona Tkachyova
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Michael B Tropak
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Alex Lee
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
| | - Alessandro Datti
- Samuel Lunenfeld Research Institute, Mount Sinai Hospital, Toronto, ON, Canada
| | - Shinya Ito
- Clinical Pharmacology and Toxicology, The Hospital for Sick Children, Toronto, ON, Canada
| | - Andreas Schulze
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, ON, Canada
- Department of Biochemistry, University of Toronto, Toronto, ON, Canada
- Department of Pediatrics, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
4
|
D'Alessio AM, Boffa I, De Stefano L, Soria LR, Brunetti-Pierri N. Liver gene transfer for metabolite detoxification in inherited metabolic diseases. FEBS Lett 2024; 598:2372-2384. [PMID: 38884367 DOI: 10.1002/1873-3468.14957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Revised: 05/28/2024] [Accepted: 06/04/2024] [Indexed: 06/18/2024]
Abstract
Inherited metabolic disorders (IMDs) are a growing group of genetic diseases caused by defects in enzymes that mediate cellular metabolism, often resulting in the accumulation of toxic substrates. The liver is a highly metabolically active organ that hosts several thousands of chemical reactions. As such, it is an organ frequently affected in IMDs. In this article, we review current approaches for liver-directed gene-based therapy aimed at metabolite detoxification in a variety of IMDs. Moreover, we discuss current unresolved challenges in gene-based therapies for IMDs.
Collapse
Affiliation(s)
- Alfonso M D'Alessio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
| | - Iolanda Boffa
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Azienda Ospedaliera Universitaria Federico II, Naples, Italy
| | - Lucia De Stefano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Leandro R Soria
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
| | - Nicola Brunetti-Pierri
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Italy
- Scuola Superiore Meridionale (SSM, School of Advanced Studies), Genomics and Experimental Medicine Program, University of Naples Federico II, Naples, Italy
- Department of Translational Medicine, Federico II University of Naples, Naples, Italy
| |
Collapse
|
5
|
Nasseri Moghaddam Z, Reinhardt EK, Thurm A, Potter BK, Smith M, Graham C, Tiller BH, Baker SA, Bilder DA, Bogar R, Britz J, Cafferty R, Coller DP, DeGrauw TJ, Hall V, Lipshutz GS, Longo N, Mercimek-Andrews S, Miller JS, Pasquali M, Salomons GS, Schulze A, Wheaton CP, Williams KF, Young SP, Li J, Balog S, Selucky T, Stockler-Ipsiroglu S, Wallis H. Establishing a Core Outcome Set for Creatine Transporter Deficiency and Guanidinoacetate Methyltransferase Deficiency. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.06.24313213. [PMID: 39371127 PMCID: PMC11451665 DOI: 10.1101/2024.09.06.24313213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
Creatine transporter (CTD) and guanidinoacetate methyltransferase (GAMT) deficiencies are rare inborn errors of creatine metabolism, resulting in cerebral creatine deficiency. Patients commonly exhibit intellectual and developmental disabilities, often accompanied by behavior problems, delayed speech, seizures, and motor impairments. There is currently no efficacious treatment for CTD, while the current management for GAMT requires lifelong treatment with a protein restricted diet and intake of high amounts of oral supplements. Efforts to develop effective, sustainable treatments for these disorders are limited by the lack of clinical and patient-derived meaningful outcomes. A core outcome set (COS) can facilitate consensus about outcomes for inclusion in studies. Unfortunately, patient and caregiver perspectives have historically been overlooked in the COS development process, thus limiting their input into the outcome selection. We partnered with caregivers and health professionals to establish the first COS for CTD and GAMT. The COS developed includes seven outcomes ("Adaptive Functioning", "Cognitive Functioning", "Emotional Dysregulation", "MRS Brain Creatine", "Seizure/Convulsions", "Expressive Communication", and "Fine Motor Functions") for both CTD and GAMT, and an additional outcome for GAMT ("Serum/Plasma Guanidinoacetate") that are important to stakeholders and consequently should be considered for measurement in every clinical trial. Caregivers were valued partners throughout the COS development process, which increased community engagement and facilitated caregiver empowerment. We expect this COS will ensure a patient-centered approach for accelerating drug development for CTD and GAMT, make clinical trial results comparable, minimize bias in clinical trial outcome selection, and promote efficient use of resources.
Collapse
Affiliation(s)
| | - Emily K. Reinhardt
- Board of Directors & Staff, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Audrey Thurm
- National Institute of Mental Health, NIH, Bethesda, MD, USA
| | - Beth K. Potter
- School of Epidemiology and Public Health, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada
| | - Maureen Smith
- Patient Partner, University of Ottawa, Ottawa, ON, Canada
| | - Celeste Graham
- Board of Directors & Staff, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Beth H. Tiller
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Steven A. Baker
- Department of Transfusion Medicine, University of Utah, Salt Lake City, UT, USA
| | - Deborah A. Bilder
- Huntsman Mental Health Institute, University of Utah, Salt Lake City, UT, USA
| | - Regina Bogar
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Jacobus Britz
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Rachel Cafferty
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Daniel P. Coller
- Board of Directors & Staff, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Ton J. DeGrauw
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Vicky Hall
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Gerald S. Lipshutz
- David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA, USA
| | - Nicola Longo
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
- David Geffen School of Medicine at the University of California-Los Angeles, Los Angeles, CA, USA
| | - Saadet Mercimek-Andrews
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Department of Medical Genetics, Faculty of Medicine & Dentistry, University of Alberta, Edmonton, AB, Canada
| | - Judith S. Miller
- Center for Autism Research, Children’s Hospital of Philadelphia, Philadelphia, PA, USA
| | - Marzia Pasquali
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
- ARUP Laboratories, Department of Pathology, University of Utah, Salt Lake City, UT, USA
| | - Gajja S. Salomons
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Laboratory Genetic Metabolic Diseases & Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, The Netherlands
| | - Andreas Schulze
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Hospital for Sick Children & University of Toronto, Toronto, ON, Canada
| | - Celine P. Wheaton
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Kayla F. Williams
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Sarah P. Young
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Division of Genetics and Metabolism, Department of Pediatrics, Duke University Medical Center, Durham, NC, USA
| | - Jasmine Li
- Department Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Sofia Balog
- Board of Directors & Staff, Association for Creatine Deficiencies, Carlsbad, CA, USA
| | | | - Sylvia Stockler-Ipsiroglu
- Department Pediatrics, University of British Columbia, Vancouver, BC, Canada
- Scientific Medical Advisory Board, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Division Biochemical Diseases, British Columbia Children’s Hospital, Vancouver, BC, Canada
| | - Heidi Wallis
- Board of Directors & Staff, Association for Creatine Deficiencies, Carlsbad, CA, USA
- Patient/Family Partner, Association for Creatine Deficiencies, Carlsbad, CA, USA
| |
Collapse
|
6
|
Couto B, Galosi S, Steel D, Kurian MA, Friedman J, Gorodetsky C, Lang AE. Severe Acute Motor Exacerbations (SAME) across Metabolic, Developmental and Genetic Disorders. Mov Disord 2024; 39:1446-1467. [PMID: 39119747 DOI: 10.1002/mds.29905] [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: 10/02/2023] [Revised: 06/08/2024] [Accepted: 06/13/2024] [Indexed: 08/10/2024] Open
Abstract
Acute presentation of severe motor disorders is a diagnostic and management challenge. We define severe acute motor exacerbations (SAME) as acute/subacute motor symptoms that persist for hours-to-days with a severity that compromise vital signs (temperature, breath, and heart rate) and bulbar function (swallowing/dysphagia). Phenomenology includes dystonia, choreoathetosis, combined movement disorders, weakness, and hemiplegic attacks. SAME can develop in diverse diseases and can be preceded by triggers or catabolic states. Recent descriptions of SAME in complex neurodevelopmental and epileptic encephalopathies have broadened appreciation of this presentation beyond inborn errors of metabolism. A high degree of clinical suspicion is required to identify appropriately targeted investigations and management. We conducted a comprehensive literature analysis of etiologies. Reported triggers are described and classified as per pathophysiological mechanism. A video of six cases displaying multiple SAME with diverse outcomes is provided. We identified 50 different conditions that manifest SAME, some associated with developmental regression. Etiologies include disorders of metabolism: energy substrate, amino acids, complex molecules, vitamins/cofactors, minerals, and neurotransmitters/synaptic vesicle cycling. Non-metabolic neurodegenerative and genetic disorders that present with movement disorders and epilepsy can additionally manifest SAME. A limited number of triggers are grouped here, together with an approach to investigations and general management strategies. Several neurogenetic and neurometabolic disorders manifest SAME. Identifying triggers can help in certain cases narrow the differential diagnosis and guide the expeditious application of targeted therapies to minimize adverse developmental and neurological consequences. This process may inform pathogenesis and eventually improve our understanding of the mechanisms that lead to the development of SAME. © 2024 International Parkinson and Movement Disorder Society.
Collapse
Affiliation(s)
- Blas Couto
- Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
- Instituto de Neurociencia Cognitiva y Traslacional, INECO-Favaloro-CONICET, Buenos Aires, Argentina
| | - Serena Galosi
- Department of Human Neuroscience, Sapienza University, Rome, Italy
| | - Dora Steel
- Molecular Neurosciences, Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| | - Manju A Kurian
- Molecular Neurosciences, Developmental Neurosciences, Zayed Centre for Research into Rare Disease in Children, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Department of Neurology, Great Ormond Street Hospital, London, United Kingdom
| | - Jennifer Friedman
- Departments of Neurosciences and Pediatrics, University of California San Diego, San Diego, California, USA
- Division of Neurology, Rady Children's Hospital; Rady Children's Institute for Genomic Medicine, San Diego, California, USA
| | - Carolina Gorodetsky
- Division of Neurology, Pediatric Deep Brain Stimulation Program, Movement Disorder and Neuromodulation Program at the Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
| | - Anthony E Lang
- Edmond J. Safra Program in Parkinson's Disease, Rossy PSP Centre and the Morton and Gloria Shulman Movement Disorders Clinic, Toronto Western Hospital, Toronto, Ontario, Canada
- Tanz Centre for Research in Neurodegenerative Disease, University of Toronto, Toronto, Ontario, Canada
- Department of Medicine, Division of Neurology, University Health Network and the University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Errichiello E, Lecca M, Vantaggiato C, Motta Z, Zanotta N, Zucca C, Bertuzzo S, Piubelli L, Pollegioni L, Bonaglia MC. Further evidence supporting the role of GTDC1 in glycine metabolism and neurodevelopmental disorders. Eur J Hum Genet 2024; 32:920-927. [PMID: 38605125 PMCID: PMC11291697 DOI: 10.1038/s41431-024-01603-0] [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: 11/29/2023] [Revised: 02/16/2024] [Accepted: 03/21/2024] [Indexed: 04/13/2024] Open
Abstract
Copy number variants (CNVs) represent the genetic cause of about 15-20% of neurodevelopmental disorders (NDDs). We identified a ~67 kb de novo intragenic deletion on chromosome 2q22.3 in a female individual showing a developmental encephalopathy characterised by epilepsy, severe intellectual disability, speech delay, microcephaly, and thin corpus callosum with facial dysmorphisms. The microdeletion involved exons 5-6 of GTDC1, encoding a putative glycosyltransferase, whose expression is particularly enriched in the nervous system. In a previous study, a balanced de novo translocation encompassing GTDC1 was reported in a male child with global developmental delay and delayed speech and language development. Based on these premises, we explored the transcriptomic profile of our proband to evaluate the functional consequences of the novel GTDC1 de novo intragenic deletion in relation to the observed neurodevelopmental phenotype. RNA-seq on the proband's lymphoblastoid cell line (LCL) showed expression changes of glycine/serine and cytokine/chemokine signalling pathways, which are related to neurodevelopment and epileptogenesis. Subsequent analysis by ELISA (enzyme-linked immunosorbent assay) and HPLC (high-performance liquid chromatography) revealed increased levels of glycine in the proband's LCL and serum compared to matched controls. Given that an increased level of glycine has been observed in the plasma samples of individuals with Rett syndrome, a condition sharing epilepsy, microcephaly, and intellectual disability with our proband, we proposed that the GTDC1 downregulation is implicated in neurodevelopmental impairment by altering glycine metabolism. Furthermore, our findings expanded the phenotypic spectrum of the novel GTDC1-related condition, including microcephaly and epilepsy among relevant clinical features.
Collapse
Affiliation(s)
- Edoardo Errichiello
- Unit of Medical Genetics, Department of Molecular Medicine, University of Pavia, Pavia, Italy.
- Neurogenetics Research Center, IRCCS Mondino Foundation, Pavia, Italy.
| | - Mauro Lecca
- Unit of Medical Genetics, Department of Molecular Medicine, University of Pavia, Pavia, Italy
| | - Chiara Vantaggiato
- Laboratory of Molecular Biology, IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Zoraide Motta
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Nicoletta Zanotta
- Unit of Clinical Neurophysiology and Epilepsy Centre, IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Claudio Zucca
- Unit of Clinical Neurophysiology and Epilepsy Centre, IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Sara Bertuzzo
- Laboratory of Cytogenetics, IRCCS E. Medea, Bosisio Parini, Lecco, Italy
| | - Luciano Piubelli
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | - Loredano Pollegioni
- Department of Biotechnology and Life Sciences, University of Insubria, Varese, Italy
| | | |
Collapse
|
8
|
Fernandes-Pires G, Azevedo MD, Lanzillo M, Roux-Petronelli C, Binz PA, Cudalbu C, Sandi C, Tenenbaum L, Braissant O. Rescue of myocytes and locomotion through AAV2/9-2YF intracisternal gene therapy in a rat model of creatine transporter deficiency. Mol Ther Methods Clin Dev 2024; 32:101251. [PMID: 38745894 PMCID: PMC11091509 DOI: 10.1016/j.omtm.2024.101251] [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: 11/01/2023] [Accepted: 04/18/2024] [Indexed: 05/16/2024]
Abstract
Creatine deficiency syndromes (CDS), caused by mutations in GATM (AGAT), GAMT, and SLC6A8, mainly affect the central nervous system (CNS). CDS show brain creatine (Cr) deficiency, intellectual disability with severe speech delay, behavioral troubles, epilepsy, and motor dysfunction. AGAT/GAMT-deficient patients lack brain Cr synthesis but express the Cr transporter SLC6A8 at the blood-brain barrier and are thus treatable by oral supplementation of Cr. In contrast, no satisfactory treatment has been identified for Cr transporter deficiency (CTD), the most frequent of CDS. We used our Slc6a8Y389C CTD rat model to develop a new AAV2/9-2YF-driven gene therapy re-establishing the functional Slc6a8 transporter in rat CNS. We show, after intra-cisterna magna AAV2/9-2YF-Slc6a8-FLAG vector injection of postnatal day 11 pups, the transduction of Slc6a8-FLAG in cerebellum, medulla oblongata, and spinal cord as well as a partial recovery of Cr in these brain regions, together with full prevention of locomotion defaults and impairment of myocyte development observed in Slc6a8Y389 C/y male rats. While more work is needed to correct those CTD phenotypes more associated with forebrain structures, this study is the first demonstrating positive effects of an AAV-driven gene therapy on CTD and thus represents a very encouraging approach to treat the so-far untreatable CTD.
Collapse
Affiliation(s)
- Gabriella Fernandes-Pires
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Marcelo Duarte Azevedo
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neurosciences Department, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Marc Lanzillo
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Clothilde Roux-Petronelli
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Pierre-Alain Binz
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Cristina Cudalbu
- Centre d'Imagerie Biomedicale (CIBM), Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Animal Imaging and Technology, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Carmen Sandi
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Liliane Tenenbaum
- Laboratory of Cellular and Molecular Neurotherapies, Clinical Neurosciences Department, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Braissant
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| |
Collapse
|
9
|
Goldstein J, Thomas-Wilson A, Groopman E, Aggarwal V, Bianconi S, Fernandez R, Hart K, Longo N, Liang N, Reich D, Wallis H, Weaver M, Young S, Mercimek-Andrews S. ClinGen variant curation expert panel recommendations for classification of variants in GAMT, GATM and SLC6A8 for cerebral creatine deficiency syndromes. Mol Genet Metab 2024; 142:108362. [PMID: 38452609 PMCID: PMC11874059 DOI: 10.1016/j.ymgme.2024.108362] [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: 12/26/2023] [Revised: 02/26/2024] [Accepted: 02/27/2024] [Indexed: 03/09/2024]
Abstract
Cerebral creatine deficiency syndromes (CCDS) are inherited metabolic phenotypes of creatine synthesis and transport. There are two enzyme deficiencies, guanidinoacetate methyltransferase (GAMT), encoded by GAMT and arginine-glycine amidinotransferase (AGAT), encoded by GATM, which are involved in the synthesis of creatine. After synthesis, creatine is taken up by a sodium-dependent membrane bound creatine transporter (CRTR), encoded by SLC6A8, into all organs. Creatine uptake is very important especially in high energy demanding organs such as the brain, and muscle. To classify the pathogenicity of variants in GAMT, GATM, and SLC6A8, we developed the CCDS Variant Curation Expert Panel (VCEP) in 2018, supported by The Clinical Genome Resource (ClinGen), a National Institutes of Health (NIH)-funded resource. We developed disease-specific variant classification guidelines for GAMT-, GATM-, and SLC6A8-related CCDS, adapted from the American College of Medical Genetics/Association of Molecular Pathology (ACMG/AMP) variant interpretation guidelines. We applied specific variant classification guidelines to 30 pilot variants in each of the three genes that have variants associated with CCDS. Our CCDS VCEP was approved by the ClinGen Sequence Variant Interpretation Working Group (SVI WG) and Clinical Domain Oversight Committee in July 2022. We curated 181 variants including 72 variants in GAMT, 45 variants in GATM, and 64 variants in SLC6A8 and submitted these classifications to ClinVar, a public variant database supported by the National Center for Biotechnology Information. Missense variants were the most common variant type in all three genes. We submitted 32 new variants and reclassified 34 variants with conflicting interpretations. We report specific phenotype (PP4) using a points system based on the urine and plasma guanidinoacetate and creatine levels, brain magnetic resonance spectroscopy (MRS) creatine level, and enzyme activity or creatine uptake in fibroblasts ranging from PP4, PP4_Moderate and PP4_Strong. Our CCDS VCEP is one of the first panels applying disease specific variant classification algorithms for an X-linked disease. The availability of these guidelines and classifications can guide molecular genetics and genomic laboratories and health care providers to assess the molecular diagnosis of individuals with a CCDS phenotype.
Collapse
Affiliation(s)
- Jennifer Goldstein
- Department of Genetics, University of North Carolina at Chapel Hill, Chapel Hill, NC, USA
| | | | - Emily Groopman
- Children’s National Hospital, 111 Michigan Ave NW, Washington, DC, USA
| | - Vimla Aggarwal
- Department of Pathology and Cell Biology, Columbia University Irving Medical Center, New York, NY, USA
| | - Simona Bianconi
- Kaiser Permanente, Southern California Permanente Group, CA, USA
| | - Raquel Fernandez
- American College of Medical Genetics and Genomics, Bethesda, MD, USA
| | - Kim Hart
- Newborn Screening Program, Utah Public Health Laboratory, Department of Health and Human Services, Salt Lake City, UT, USA
| | - Nicola Longo
- Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | | | - Daniel Reich
- Newborn Screening Program, Utah Public Health Laboratory, Department of Health and Human Services, Salt Lake City, UT, USA
| | - Heidi Wallis
- Association for Creatine Deficiencies, Carlsbad, CA, USA
| | - Meredith Weaver
- American College of Medical Genetics and Genomics, Bethesda, MD, USA
| | - Sarah Young
- Department of Pediatrics, Duke University School of Medicine, Durham, NC, USA
| | | |
Collapse
|
10
|
Marten LM, Krätzner R, Salomons GS, Fernandez Ojeda M, Dechent P, Gärtner J, Huppke P, Dreha-Kulaczewski S. Long term follow-up in GAMT deficiency - Correlation of therapy regimen, biochemical and in vivo brain proton MR spectroscopy data. Mol Genet Metab Rep 2024; 38:101053. [PMID: 38469086 PMCID: PMC10926185 DOI: 10.1016/j.ymgmr.2024.101053] [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: 11/04/2023] [Revised: 01/11/2024] [Accepted: 01/12/2024] [Indexed: 03/13/2024] Open
Abstract
GAMT deficiency is a rare autosomal recessive disease within the group of cerebral creatine deficiency syndromes. Cerebral creatine depletion and accumulation of guanidinoacetate (GAA) lead to clinical presentation with intellectual disability, seizures, speech disturbances and movement disorders. Treatment consists of daily creatine supplementation to increase cerebral creatine, reduction of arginine intake and supplementation of ornithine for reduction of toxic GAA levels. This study represents the first long-term follow-up over a period of 14 years, with detailed clinical data, biochemical and multimodal neuroimaging findings. Developmental milestones, brain MRI, quantitative single voxel 1H magnetic resonance spectroscopy (MRS) and biochemical analyses were assessed. The results reveal insights into the dose dependent effects of creatine/ornithine supplementation and expand the phenotypic spectrum of GAMT deficiency. Of note, the creatine concentrations, which were regularly monitored over a long follow-up period, increased significantly over time, but did not reach age matched control ranges. Our patient is the second reported to show normal neurocognitive outcome after an initial delay, stressing the importance of early diagnosis and treatment initiation.
Collapse
Affiliation(s)
- Lara M. Marten
- Department of Pediatrics and Adolescent Medicine, University Medical Center Goettingen, Germany
| | - Ralph Krätzner
- Department of Pediatrics and Adolescent Medicine, University Medical Center Goettingen, Germany
| | - Gajja S. Salomons
- Amsterdam UMC location University of Amsterdam, Dept of Laboratory Medicine, Laboratory Genetic Metabolic Diseases and Dept of Pediatrics Emma Children's Hospital, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Matilde Fernandez Ojeda
- Amsterdam UMC location University of Amsterdam, Dept of Laboratory Medicine, Laboratory Genetic Metabolic Diseases and Dept of Pediatrics Emma Children's Hospital, Meibergdreef 9, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Amsterdam, the Netherlands
| | - Peter Dechent
- MR-Research in Neurosciences, Department of Cognitive Neurology, University Medical Center Goettingen, Germany
| | - Jutta Gärtner
- Department of Pediatrics and Adolescent Medicine, University Medical Center Goettingen, Germany
| | - Peter Huppke
- Department of Neuropediatrics, Jena University Hospital, Germany
| | | |
Collapse
|
11
|
Yıldız Y, Ardıçlı D, Göçmen R, Yalnızoğlu D, Topçu M, Coşkun T, Tokatlı A, Haliloğlu G. Electro-clinical features and long-term outcomes in guanidinoacetate methyltransferase (GAMT) deficiency. Eur J Paediatr Neurol 2024; 49:66-72. [PMID: 38394710 DOI: 10.1016/j.ejpn.2024.02.006] [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: 11/21/2023] [Revised: 02/06/2024] [Accepted: 02/11/2024] [Indexed: 02/25/2024]
Abstract
OBJECTIVE To evaluate clinical characteristics and long-term outcomes in patients with guanidinoacetate methyltransferase (GAMT) deficiency with a special emphasis on seizures and electroencephalography (EEG) findings. METHODS We retrospectively analyzed the clinical and molecular characteristics, seizure types, EEG findings, neuroimaging features, clinical severity scores, and treatment outcomes in six patients diagnosed with GAMT deficiency. RESULTS Median age at presentation and diagnosis were 11.5 months (8-12 months) and 63 months (18 months -11 years), respectively. Median duration of follow-up was 14 years. Global developmental delay (6/6) and seizures (5/6) were the most common symptoms. Four patients presented with febrile seizures. The age at seizure-onset ranged between 8 months and 4 years. Most common seizure types were generalized tonic seizures (n = 4) and motor seizures resulting in drop attacks (n = 3). Slow background activity (n = 5) and generalized irregular sharp and slow waves (n = 3) were the most common EEG findings. Burst-suppression and electrical status epilepticus during slow-wave sleep (ESES) pattern was present in one patient. Three of six patients had drug-resistant epilepsy. Post-treatment clinical severity scores showed improvement regarding movement disorders and epilepsy. All patients were seizure-free in the follow-up. CONCLUSIONS Epilepsy is one of the main symptoms in GAMT deficiency with various seizure types and non-specific EEG findings. Early diagnosis and initiation of treatment are crucial for better seizure and cognitive outcomes. This long-term follow up study highlights to include cerebral creatine deficiency syndromes in the differential diagnosis of patients with global developmental delay and epilepsy and describes the course under treatment.
Collapse
Affiliation(s)
- Yılmaz Yıldız
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Metabolism and Nutrition, Turkey.
| | - Didem Ardıçlı
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Neurology, Turkey
| | - Rahşan Göçmen
- Hacettepe University Faculty of Medicine, Department of Radiology, Turkey.
| | - Dilek Yalnızoğlu
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Neurology, Turkey.
| | - Meral Topçu
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Neurology, Turkey
| | - Turgay Coşkun
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Metabolism and Nutrition, Turkey
| | - Ayşegül Tokatlı
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Metabolism and Nutrition, Turkey.
| | - Göknur Haliloğlu
- Hacettepe University Faculty of Medicine, Department of Pediatrics, Division of Pediatric Neurology, Turkey.
| |
Collapse
|
12
|
Latzer IT, Pearl PL. Treatable inherited metabolic epilepsies. Epilepsy Behav 2024; 151:109621. [PMID: 38237465 DOI: 10.1016/j.yebeh.2024.109621] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 12/30/2023] [Accepted: 01/02/2024] [Indexed: 02/09/2024]
Abstract
Inherited metabolic epilepsies (IMEs) represent inherited metabolic disorders predominately presenting with seizures. While most IMEs are currently managed with symptomatic and supportive therapies, some are amenable to disorder-specific targeted treatments. In most cases, these treatments are effective only if given in a narrow time window early in the lives of affected patients. Hence, prompt recognition of treatable inherited metabolic epilepsies at an early age and as soon as symptoms appear has paramount importance. Herein, we provide an overview of inherited metabolic epilepsies, which presently have established targeted treatments showing clinical efficacy in reducing seizure burden and improving neurodevelopmental outcomes. These therapeutic modalities range from specific diets, vitamins, and supplementation of organic compounds to synthetic pharmacological agents and novel genetic-based therapies that alter the biochemical pathways of these disorders at the cellular or molecular level, steering them to their normal function.
Collapse
Affiliation(s)
- Itay Tokatly Latzer
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA; Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
| | - Phillip L Pearl
- Department of Neurology, Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Bian X, Zhu J, Jia X, Liang W, Yu S, Li Z, Zhang W, Rao Y. Suggestion of creatine as a new neurotransmitter by approaches ranging from chemical analysis and biochemistry to electrophysiology. eLife 2023; 12:RP89317. [PMID: 38126335 PMCID: PMC10735228 DOI: 10.7554/elife.89317] [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] [Indexed: 12/23/2023] Open
Abstract
The discovery of a new neurotransmitter, especially one in the central nervous system, is both important and difficult. We have been searching for new neurotransmitters for 12 y. We detected creatine (Cr) in synaptic vesicles (SVs) at a level lower than glutamate and gamma-aminobutyric acid but higher than acetylcholine and 5-hydroxytryptamine. SV Cr was reduced in mice lacking either arginine:glycine amidinotransferase (a Cr synthetase) or SLC6A8, a Cr transporter with mutations among the most common causes of intellectual disability in men. Calcium-dependent release of Cr was detected after stimulation in brain slices. Cr release was reduced in Slc6a8 and Agat mutants. Cr inhibited neocortical pyramidal neurons. SLC6A8 was necessary for Cr uptake into synaptosomes. Cr was found by us to be taken up into SVs in an ATP-dependent manner. Our biochemical, chemical, genetic, and electrophysiological results are consistent with the possibility of Cr as a neurotransmitter, though not yet reaching the level of proof for the now classic transmitters. Our novel approach to discover neurotransmitters is to begin with analysis of contents in SVs before defining their function and physiology.
Collapse
Affiliation(s)
- Xiling Bian
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
| | - Jiemin Zhu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
| | - Xiaobo Jia
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
| | - Wenjun Liang
- Chinese Institutes of Medical Research, Capital Medical UniversityBeijingChina
- Changping Laboratory, Yard 28, Science Park Road, Changping DistrictBeijingChina
| | - Sihan Yu
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Changping Laboratory, Yard 28, Science Park Road, Changping DistrictBeijingChina
| | - Zhiqiang Li
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
| | - Wenxia Zhang
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institutes of Medical Research, Capital Medical UniversityBeijingChina
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
| | - Yi Rao
- Laboratory of Neurochemical Biology, PKU-IDG/McGovern Institute for Brain Research, Peking-Tsinghua Center for Life Sciences, School of Life Sciences, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Department of Molecular and Cellular Pharmacology, School of Pharmaceutical Sciences, Peking UniversityBeijingChina
- Chinese Institute for Brain Research (CIBR)BeijingChina
- Chinese Institutes of Medical Research, Capital Medical UniversityBeijingChina
- Changping Laboratory, Yard 28, Science Park Road, Changping DistrictBeijingChina
- Institute of Molecular Physiology, Shenzhen Bay LaboratoryShenzhenChina
- Research Unit of Medical Neurobiology, Chinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
14
|
Tropak MB, Tkachyova I, Gu R, Lee A, Schulze A. Evidence of an intracellular creatine-sensing mechanism that modulates creatine biosynthesis via AGAT expression in human HAP1 cells. Sci Rep 2023; 13:22392. [PMID: 38104212 PMCID: PMC10725494 DOI: 10.1038/s41598-023-49860-1] [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: 08/10/2023] [Accepted: 12/12/2023] [Indexed: 12/19/2023] Open
Abstract
Cellular homeostasis of creatine (CT), integral part of the energy buffering and transducing system connecting intracellular sites of ATP production and utilization, comprises of mechanisms that increase CT, i.e., biosynthesis and cellular uptake, and CT-lowering processes, such as export and non-enzymatic conversion to creatinine. The biosynthesis of CT is controlled by negative feedback loop via suppression of the rate-limiting enzyme arginine:glycine amidinotransferase (AGAT). Although the regulatory mechanism involved is not well understood, AGAT suppression is successfully used in patients with guanidinoacetate methyltransferase (GAMT) deficiency to reduce the neurotoxic accumulation of the AGAT-mediated guanidinoacetate production by supplementing patients with CT. Utilizing the CT-dependent feedback loop for the upregulation of AGAT expression may well represent a therapeutic target for an additional CT deficiency syndrome, the CT transporter (CrT) defect, for which no effective treatment option is available so far. We have used CRISPR to tag the C-terminus of AGAT with a nanoluc luciferase (NLuc) reporter in HAP1 cells. A biphasic decay of AGAT-NLuc in response to increasing extracellular CT was observed, whereas the decrease in AGAT-NLuc expression was directly proportional to the rise in intracellular CT levels with an approximate IC50 of 1-2 mM. CRISPR generated HAP1 CrT null cells and HAP1 CrT null cells stably expressing a CrT-GFP fusion protein further demonstrated that the biphasic response to extracellular CT is mediated by a high-affinity (Km 9-10 µM) CrT dependent, saturable mechanism and a CrT independent, unsaturable uptake process. The direct response to intracellular CT suggests the existence of an intracellular CT sensing system enabling a dynamic cell response to changing CT concentration that is relevant for cellular CT homeostasis.
Collapse
Affiliation(s)
- Michael B Tropak
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Ilona Tkachyova
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
| | - Ray Gu
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Alex Lee
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada
- Department of Biochemistry, University of Toronto, Toronto, Canada
| | - Andreas Schulze
- Genetics and Genome Biology, The Hospital for Sick Children, Toronto, Canada.
- Department of Pediatrics, University of Toronto, Toronto, Canada.
- Department of Biochemistry, University of Toronto, Toronto, Canada.
| |
Collapse
|
15
|
Libell JL, Lakhani DA, Balar AB, Khan M, Carpenter JS, Joseph JT. Guanidinoacetate N-methyltransferase deficiency: Case report and brief review of the literature. Radiol Case Rep 2023; 18:4331-4337. [PMID: 37808418 PMCID: PMC10550807 DOI: 10.1016/j.radcr.2023.09.026] [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: 08/11/2023] [Revised: 08/29/2023] [Accepted: 09/05/2023] [Indexed: 10/10/2023] Open
Abstract
Guanidinoacetate N-methyltransferase (GAMT) deficiency is a rare autosomal recessive disorder characterized by a decrease in creatine synthesis, resulting in cerebral creatine deficiency syndrome (CCDS). GAMT deficiency is caused by mutations in the GAMT gene located on chromosome 19, which impairs the conversion of guanidinoacetic acid (GAA) to creatine. The resulting accumulation of the toxic metabolite GAA and the lack of creatine lead to various symptoms, including global developmental delays, behavioral issues, and epilepsy. The gold standard for diagnosis of GAMT deficiency is genetic testing. Treatment options for GAMT deficiency include creatine supplementation, ornithine supplementation, arginine restriction, and sodium benzoate supplementation. These treatment options have been shown to improve movement disorders and epileptic symptoms, but their impact on intellectual and speech development is limited. Early intervention has shown promising results in normalizing neurological development in a minor subgroup of patients. Therefore, there is a growing need for newborn screening techniques to detect GAMT deficiency early and prevent permanent neurological delays. Here we report a case of GAMT deficiency with emphasis on imaging presentation. Our case showed reduced brain parenchyma creatine stores on MR Spectroscopy, which may provide an avenue to aid in early diagnosis.
Collapse
Affiliation(s)
- Joshua L. Libell
- School of Medicine, West Virginia University, Morgantown, WV, 26506, USA
| | - Dhairya A. Lakhani
- Department of Radiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26506, USA
| | - Aneri B. Balar
- Department of Radiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26506, USA
| | - Musharaf Khan
- Department of Radiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26506, USA
| | - Jeffrey S. Carpenter
- Department of Radiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26506, USA
| | - Joe T. Joseph
- Department of Radiology, West Virginia University, 1 Medical Center Dr, Morgantown, WV 26506, USA
| |
Collapse
|
16
|
Lyons EL, Watson D, Alodadi MS, Haugabook SJ, Tawa GJ, Hannah-Shmouni F, Porter FD, Collins JR, Ottinger EA, Mudunuri US. Rare disease variant curation from literature: assessing gaps with creatine transport deficiency in focus. BMC Genomics 2023; 24:460. [PMID: 37587458 PMCID: PMC10433598 DOI: 10.1186/s12864-023-09561-5] [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: 04/04/2023] [Accepted: 08/08/2023] [Indexed: 08/18/2023] Open
Abstract
BACKGROUND Approximately 4-8% of the world suffers from a rare disease. Rare diseases are often difficult to diagnose, and many do not have approved therapies. Genetic sequencing has the potential to shorten the current diagnostic process, increase mechanistic understanding, and facilitate research on therapeutic approaches but is limited by the difficulty of novel variant pathogenicity interpretation and the communication of known causative variants. It is unknown how many published rare disease variants are currently accessible in the public domain. RESULTS This study investigated the translation of knowledge of variants reported in published manuscripts to publicly accessible variant databases. Variants, symptoms, biochemical assay results, and protein function from literature on the SLC6A8 gene associated with X-linked Creatine Transporter Deficiency (CTD) were curated and reported as a highly annotated dataset of variants with clinical context and functional details. Variants were harmonized, their availability in existing variant databases was analyzed and pathogenicity assignments were compared with impact algorithm predictions. 24% of the pathogenic variants found in PubMed articles were not captured in any database used in this analysis while only 65% of the published variants received an accurate pathogenicity prediction from at least one impact prediction algorithm. CONCLUSIONS Despite being published in the literature, pathogenicity data on patient variants may remain inaccessible for genetic diagnosis, therapeutic target identification, mechanistic understanding, or hypothesis generation. Clinical and functional details presented in the literature are important to make pathogenicity assessments. Impact predictions remain imperfect but are improving, especially for single nucleotide exonic variants, however such predictions are less accurate or unavailable for intronic and multi-nucleotide variants. Developing text mining workflows that use natural language processing for identifying diseases, genes and variants, along with impact prediction algorithms and integrating with details on clinical phenotypes and functional assessments might be a promising approach to scale literature mining of variants and assigning correct pathogenicity. The curated variants list created by this effort includes context details to improve any such efforts on variant curation for rare diseases.
Collapse
Affiliation(s)
- Erica L Lyons
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Daniel Watson
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Mohammad S Alodadi
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Sharie J Haugabook
- Division of Preclinical Innovation, Therapeutic Development Branch, Therapeutics for Rare and Neglected Diseases (TRND) Program, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Gregory J Tawa
- Division of Preclinical Innovation, Therapeutic Development Branch, Therapeutics for Rare and Neglected Diseases (TRND) Program, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Fady Hannah-Shmouni
- Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Forbes D Porter
- Division of Translational Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Jack R Collins
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA
| | - Elizabeth A Ottinger
- Division of Preclinical Innovation, Therapeutic Development Branch, Therapeutics for Rare and Neglected Diseases (TRND) Program, National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Uma S Mudunuri
- Advanced Biomedical Computational Science, Frederick National Laboratory for Cancer Research, Frederick, MD, 21702, USA.
| |
Collapse
|
17
|
Ream MA, Lam WK, Grosse SD, Ojodu J, Jones E, Prosser LA, Rose AM, Comeau AM, Tanksley S, Powell CM, Kemper AR. Evidence and Recommendation for Guanidinoacetate Methyltransferase Deficiency Newborn Screening. Pediatrics 2023; 152:e2023062100. [PMID: 37465909 PMCID: PMC10527896 DOI: 10.1542/peds.2023-062100] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/03/2023] [Indexed: 07/20/2023] Open
Abstract
Guanidinoacetate methyltransferase (GAMT) deficiency is an autosomal recessive disorder of creatine biosynthesis due to pathogenic variants in the GAMT gene that lead to cerebral creatine deficiency and neurotoxic levels of guanidinoacetate. Untreated, GAMT deficiency is associated with hypotonia, significant intellectual disability, limited speech development, recurrent seizures, behavior problems, and involuntary movements. The birth prevalence of GAMT deficiency is likely between 0.5 and 2 per million live births. On the basis of small case series and sibling data, presymptomatic treatment with oral supplements of creatine, ornithine, and sodium benzoate, and a protein-restricted diet to reduce arginine intake, appear to substantially improve health and developmental outcomes. Without newborn screening, diagnosis typically happens after the development of significant impairment, when treatment has limited utility. GAMT deficiency newborn screening can be incorporated into the tandem-mass spectrometry screening that is already routinely used for newborn screening, with about 1 per 100 000 newborns screening positive. After a positive screen, diagnosis is established by finding an elevated guanidinoacetate concentration and low creatine concentration in the blood. Although GAMT deficiency is significantly more rare than other conditions included in newborn screening, the feasibility of screening, the low number of positive results, the relative ease of diagnosis, and the expected benefit of presymptomatic dietary therapy led to a recommendation from the Advisory Committee on Heritable Disorders in Newborns and Children to the Secretary of Health and Human Services that GAMT deficiency be added to the Recommended Uniform Screening Panel. This recommendation was accepted in January 2023.
Collapse
Affiliation(s)
- Margie A. Ream
- Division of Child Neurology, Nationwide Children’s Hospital, Columbus, Ohio
| | - Wendy K.K. Lam
- Duke Clinical and Translational Science Institute, Duke University School of Medicine, Durham, North Carolina
| | - Scott D. Grosse
- National Center on Birth Defects and Developmental Disabilities, Centers for Disease Control and Prevention, Atlanta, Georgia
| | - Jelili Ojodu
- Association of Public Health Laboratories, Silver Spring, Maryland
| | - Elizabeth Jones
- Association of Public Health Laboratories, Silver Spring, Maryland
| | - Lisa A. Prosser
- Susan B. Meister Child Health Evaluation and Research Center, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Angela M. Rose
- Susan B. Meister Child Health Evaluation and Research Center, Department of Pediatrics, University of Michigan, Ann Arbor, Michigan
| | - Anne Marie Comeau
- New England Newborn Screening Program, Department of Pediatrics, UMass Chan School of Medicine, Worcester, Massachusetts
| | - Susan Tanksley
- Texas Department of State Health Services, Laboratory Services Section, Austin, Texas
| | - Cynthia M. Powell
- Division of Genetics and Metabolism, Department of Pediatrics, School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Alex R. Kemper
- Division of Primary Care Pediatrics, Nationwide Children’s Hospital, Columbus, Ohio
| |
Collapse
|
18
|
Bianchi M, Rossi L, Pierigè F, Biagiotti S, Bregalda A, Tasini F, Magnani M. Preclinical and clinical developments in enzyme-loaded red blood cells: an update. Expert Opin Drug Deliv 2023; 20:921-935. [PMID: 37249524 DOI: 10.1080/17425247.2023.2219890] [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: 02/01/2023] [Accepted: 05/26/2023] [Indexed: 05/31/2023]
Abstract
INTRODUCTION We have previously described the preclinical developments in enzyme-loaded red blood cells to be used in the treatment of several rare diseases, as well as in chronic conditions. AREA COVERED Since our previous publication we have seen further progress in the previously discussed approaches and, interestingly enough, in additional new studies that further strengthen the idea that red blood cell-based therapeutics may have unique advantages over conventional enzyme replacement therapies in terms of efficacy and safety. Here we highlight these investigations and compare, when possible, the reported results versus the current therapeutic approaches. EXPERT OPINION The continuous increase in the number of new potential applications and the progress from the encapsulation of a single enzyme to the engineering of an entire metabolic pathway open the field to unexpected developments and confirm the role of red blood cells as cellular bioreactors that can be conveniently manipulated to acquire useful therapeutic metabolic abilities. Positioning of these new approaches versus newly approved drugs is essential for the successful transition of this technology from the preclinical to the clinical stage and hopefully to final approval.
Collapse
Affiliation(s)
- Marzia Bianchi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Luigia Rossi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- EryDel SpA, Bresso, MI, Italy
| | - Francesca Pierigè
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Sara Biagiotti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alessandro Bregalda
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Filippo Tasini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mauro Magnani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- EryDel SpA, Bresso, MI, Italy
| |
Collapse
|
19
|
Meera P, Uusi-Oukari M, Wallner M, Lipshutz GS. Guanidinoacetate (GAA) is a potent GABA A receptor GABA mimetic: Implications for neurological disease pathology. J Neurochem 2023; 165:445-454. [PMID: 36726215 DOI: 10.1111/jnc.15774] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2022] [Revised: 01/24/2023] [Accepted: 01/28/2023] [Indexed: 02/03/2023]
Abstract
Impairment of excretion and enzymatic processing of nitrogen, for example, because of liver or kidney failure, or with urea cycle and creatine synthesis enzyme defects, surprisingly leads to primarily neurologic symptoms, yet the exact mechanisms remain largely mysterious. In guanidinoacetate N-methyltransferase (GAMT) deficiency, the guanidino compound guanidinoacetate (GAA) increases dramatically, including in the cerebrospinal fluid (CSF), and has been implicated in mediating the neurological symptoms in GAMT-deficient patients. GAA is synthesized by arginine-glycine amidinotransferase (AGAT), a promiscuous enzyme that not only transfers the amidino group from arginine to glycine, but also to primary amines in, for example, GABA and taurine to generate γ-guanidinobutyric acid (γ-GBA) and guanidinoethanesulfonic acid (GES), respectively. We show that GAA, γ-GBA, and GES share structural similarities with GABA, evoke GABAA receptor (GABAA R) mediated currents (whereas creatine [methylated GAA] and arginine failed to evoke discernible currents) in cerebellar granule cells in mouse brain slices and displace the high-affinity GABA-site radioligand [3 H]muscimol in total brain homogenate GABAA Rs. While γ-GBA and GES are GABA agonists and displace [3 H]muscimol (EC50 /IC50 between 10 and 40 μM), GAA stands out as particularly potent in both activating GABAA Rs (EC50 ~6 μM) and also displacing the GABAA R ligand [3 H]muscimol (IC50 ~3 μM) at pathophysiologically relevant concentrations. These findings stress the role of substantially elevated GAA as a primary neurotoxic agent in GAMT deficiency and we discuss the potential role of GAA in arginase (and creatine transporter) deficiency which show a much more modest increase in GAA concentrations yet share the unique hyperexcitability neuropathology with GAMT deficiency. We conclude that orthosteric activation of GABAA Rs by GAA, and potentially other GABAA R mimetic guanidino compounds (GCs) like γ-GBA and GES, interferes with normal inhibitory GABAergic neurotransmission which could mediate, and contribute to, neurotoxicity.
Collapse
Affiliation(s)
- Pratap Meera
- Department of Neurobiology, University of California, Los Angeles, California, USA
| | - Mikko Uusi-Oukari
- Integrative Physiology and Pharmacology, Institute of Biomedicine, University of Turku, Turku, Finland
| | - Martin Wallner
- Departments of Surgery, University of California, Los Angeles, California, USA
| | - Gerald S Lipshutz
- Departments of Surgery, University of California, Los Angeles, California, USA.,Molecular & Medical Pharmacology, University of California, Los Angeles, California, USA.,Intellectual and Developmental Disabilities Research Center, University of California, Los Angeles, California, USA.,Semel Institute for Neuroscience, University of California, Los Angeles, California, USA
| |
Collapse
|
20
|
Aghamollaii V, Meshkat S, Bakhtiari S, Alehabib E, Firouzabadi SG, Molaei S, Kruer MC, Darvish H. Guanidinoacetate Methyltransferase Deficiency, a Treatable Neurodevelopmental Disorder. JOURNAL OF PEDIATRIC EPILEPSY 2023. [DOI: 10.1055/s-0042-1760291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
AbstractGuanidinoacetate methyltransferase (GAMT) deficiency is an autosomal recessive inborn error of creatine synthesis that results in intellectual disability, epilepsy, expressive language delay, and dystonia. We report data from two siblings with an uncommon GAMT deficiency phenotype and their clinical, biochemical, imaging, and treatment findings. The older sibling had intellectual disability, epilepsy, and generalized dystonia. The younger sibling had intellectual disability and generalized dystonia. After treatment with creatine, verbal fluency improved, as well as dystonia and aggression. This study confirms that in patients with unexplained intellectual disability, epilepsy, and/or movement disorders, GAMT deficiency should be considered. GAMT-associated cerebral creatine deficiency syndrome is a potentially treatable condition and can be identified by elevated levels of guanidinoacetate in plasma or urine or by a significantly decreased creatine peak on magnetic resonance spectroscopy.
Collapse
Affiliation(s)
- Vajiheh Aghamollaii
- Neurology Department, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Shakila Meshkat
- Neurology Department, Roozbeh Psychiatric Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | - Somayeh Bakhtiari
- Barrow Neurological Institute, Phoenix Children's Hospital, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, United States
| | - Elham Alehabib
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | | | - Samira Molaei
- Student Research Committee, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Michael C. Kruer
- Barrow Neurological Institute, Phoenix Children's Hospital, University of Arizona College of Medicine – Phoenix, Phoenix, Arizona, United States
| | - Hossein Darvish
- Cancer Research Center, Department of Medical Genetics, School of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| |
Collapse
|
21
|
Expanding the neuroimaging findings of guanidinoacetate methyltransferase deficiency in an Iranian girl with a homozygous frameshift variant in the GAMT. Neurogenetics 2023; 24:67-78. [PMID: 36633690 DOI: 10.1007/s10048-022-00708-2] [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: 09/09/2022] [Accepted: 12/28/2022] [Indexed: 01/13/2023]
Abstract
Guanidinoacetate methyltransferase deficiency (GAMTD) is a treatable neurodevelopmental disorder with normal or nonspecific imaging findings. Here, we reported a 14-month-old girl with GAMTD and novel findings on brain magnetic resonance imaging (MRI).A 14-month-old female patient was referred to Myelin Disorders Clinic due to onset of seizures and developmental regression following routine vaccination at 4 months of age. Brain MRI, prior to initiation of treatment, showed high signal intensity in T2-weighted imaging in bilateral thalami, globus pallidus, subthalamic nuclei, substantia nigra, dentate nuclei, central tegmental tracts in the brainstem, and posterior periventricular white matter which was masquerading for mitochondrial leukodystrophy. Basic metabolic tests were normal except for low urine creatinine; however, exome sequencing identified a homozygous frameshift deletion variant [NM_000156: c.491del; (p.Gly164AlafsTer14)] in the GAMT. Biallelic pathogenic or likely pathogenic variants cause GAMTD. We confirmed the homozygous state for this variant in the proband, as well as the heterozygote state in the parents by Sanger sequencing.MRI features in GAMTD can mimic mitochondrial leukodystrophy. Pediatric neurologists should be aware of variable MRI findings in GAMTD since they would be misleading to other diagnoses.
Collapse
|
22
|
Almaghrabi MA, Muthaffar OY, Alahmadi SA, Abdulsbhan MA, Bamusa M, Aljezani MA, Bahowarth SY, Alyazidi AS, Aggad WS. GAMT Deficiency Among Pediatric Population: Clinical and Molecular Characteristics and Management. Child Neurol Open 2023; 10:2329048X231215630. [PMID: 38020815 PMCID: PMC10655665 DOI: 10.1177/2329048x231215630] [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: 08/29/2023] [Revised: 10/12/2023] [Accepted: 11/03/2023] [Indexed: 12/01/2023] Open
Abstract
Objective: Analyze the treatment modalities used in real practice by synthesizing available literature. Methods: We reviewed and evaluated 52 cases of GAMT deficiency including 4 novel cases from Saudi Arabia diagnosed using whole-exome sequencing. All data utilized graphical presentation in the form of line charts and illustrated graphs. Results: The mean current age of was 117 months (±29.03) (range 12-372 months). The mean age of disease onset was 28.32 months (±13.68) (range 8 days - 252 months). The most prevalent symptom was developmental delays, mainly speech and motor, seizures, and intellectual disability. The male-to-female ratio was 3:1. Multiple treatments were used, with 54 pharmacological interventions, valproic acid being the most common. Creatinine monohydrate was the prevalent dietary intervention, with 25 patients reporting an improvement. Conclusion: The study suggests that efficient treatment with appropriate dietary intervention can improve patients' health, stressing that personalized treatment programs are essential in managing this disorder.
Collapse
Affiliation(s)
- Majdah A. Almaghrabi
- Division of Pediatrics Neurology, Department of Pediatrics, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Osama Y. Muthaffar
- Division of Pediatrics Neurology, Department of Pediatrics, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Sereen A. Alahmadi
- Division of Pediatrics Neurology, Department of Pediatrics, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mashael A. Abdulsbhan
- Division of Pediatrics Neurology, Department of Pediatrics, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mashael Bamusa
- Division of Pediatrics Neurology, Department of Pediatrics, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Maram Ahmed Aljezani
- Division of Pediatrics Neurology, Department of Pediatrics, King Abdulaziz University, Jeddah, Saudi Arabia
| | | | - Anas S. Alyazidi
- Faculty of Medicine, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Waheeb S. Aggad
- Department of Anatomy, Faculty of Medicine, University of Jeddah, Jeddah, Saudi Arabia
| |
Collapse
|
23
|
Ataxia in Neurometabolic Disorders. Metabolites 2022; 13:metabo13010047. [PMID: 36676973 PMCID: PMC9866741 DOI: 10.3390/metabo13010047] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 12/19/2022] [Accepted: 12/19/2022] [Indexed: 12/29/2022] Open
Abstract
Ataxia is a movement disorder that manifests during the execution of purposeful movements. It results from damage to the structures of the cerebellum and its connections or the posterior cords of the spinal cord. It should be noted that, in addition to occurring as part of many diseases, pediatric ataxia is a common symptom in neurometabolic diseases. To date, there are more than 150 inherited metabolic disorders that can manifest as ataxia in children. Neuroimaging studies (magnetic resonance imaging of the head and spinal cord) are essential in the diagnosis of ataxia, and genetic studies are performed when metabolic diseases are suspected. It is important to remember that most of these disorders are progressive if left untreated. Therefore, it is crucial to include neurometabolic disorders in the differential diagnosis of ataxia, so that an early diagnosis can be made. Initiating prompt treatment influences positive neurodevelopmental outcomes.
Collapse
|
24
|
Liu N, Sun Q. Laboratory Diagnosis of Cerebral Creatine Deficiency Syndromes by Determining Creatine and Guanidinoacetate in Plasma and Urine. Methods Mol Biol 2022; 2546:129-140. [PMID: 36127584 DOI: 10.1007/978-1-0716-2565-1_12] [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] [Indexed: 11/30/2022]
Abstract
Cerebral creatine deficiency syndromes are caused by the dysfunctional creatine biosynthesis or transport and comprise three hereditary neurodevelopmental defects including arginine-glycine amidinotransferase (AGAT), guanidinoacetate methyltransferase (GAMT), and creatine transporter deficiencies. All conditions are characterized by seizures, intellectual disability, and behavioral abnormalities. Laboratory diagnosis of these disorders relies on the determination of creatine and guanidinoacetate concentrations in both plasma and urine. Here we describe a rapid quantitative UPLC/MS/MS method for the simultaneous determination of these analytes using a normal-phase HILIC column after analyte derivatization. The approach is suitable for neonatal screening follow-ups and monitoring of the treatment for creatine deficiency syndromes.
Collapse
Affiliation(s)
- Ning Liu
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Baylor Genetics, Houston, TX, USA
| | - Qin Sun
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA.
- Baylor Genetics, Houston, TX, USA.
| |
Collapse
|
25
|
Stezin A, Pal PK. Treatable Ataxias: How to Find the Needle in the Haystack? J Mov Disord 2022; 15:206-226. [PMID: 36065614 DOI: 10.14802/jmd.22069] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Accepted: 07/05/2022] [Indexed: 11/24/2022] Open
Abstract
Treatable ataxias are a group of ataxic disorders with specific treatments. These disorders include genetic and metabolic disorders, immune-mediated ataxic disorders, and ataxic disorders associated with infectious and parainfectious etiology, vascular causes, toxins and chemicals, and endocrinopathies. This review provides a comprehensive overview of different treatable ataxias. The major metabolic and genetic treatable ataxic disorders include ataxia with vitamin E deficiency, abetalipoproteinemia, cerebrotendinous xanthomatosis, Niemann-Pick disease type C, autosomal recessive cerebellar ataxia due to coenzyme Q10 deficiency, glucose transporter type 1 deficiency, and episodic ataxia type 2. The treatment of these disorders includes the replacement of deficient cofactors and vitamins, dietary modifications, and other specific treatments. Treatable ataxias with immune-mediated etiologies include gluten ataxia, anti-glutamic acid decarboxylase antibody-associated ataxia, steroid-responsive encephalopathy associated with autoimmune thyroiditis, Miller-Fisher syndrome, multiple sclerosis, and paraneoplastic cerebellar degeneration. Although dietary modification with a gluten-free diet is adequate in gluten ataxia, other autoimmune ataxias are managed by short-course steroids, plasma exchange, or immunomodulation. For autoimmune ataxias secondary to malignancy, treatment of tumor can reduce ataxic symptoms. Chronic alcohol consumption, antiepileptics, anticancer drugs, exposure to insecticides, heavy metals, and recreational drugs are potentially avoidable and treatable causes of ataxia. Infective and parainfectious causes of cerebellar ataxias include acute cerebellitis, postinfectious ataxia, Whipple's disease, meningoencephalitis, and progressive multifocal leukoencephalopathy. These disorders are treated with steroids and antibiotics. Recognizing treatable disorders is of paramount importance when dealing with ataxias given that early treatment can prevent permanent neurological sequelae.
Collapse
Affiliation(s)
- Albert Stezin
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India.,Centre for Brain Research, Indian Institute of Science, Bengaluru, India
| | - Pramod Kumar Pal
- Department of Neurology, National Institute of Mental Health and Neurosciences (NIMHANS), Bengaluru, India
| |
Collapse
|
26
|
Burns W, Chaudhari BP, Haffner DN. Neurogenetic and Metabolic Mimics of Common Neonatal Neurological Disorders. Semin Pediatr Neurol 2022; 42:100972. [PMID: 35868729 DOI: 10.1016/j.spen.2022.100972] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 04/05/2022] [Accepted: 04/05/2022] [Indexed: 10/18/2022]
Abstract
Neurogenetic and metabolic diseases often present in the neonatal period, masquerading as other disorders, most commonly as neonatal encephalopathy and seizures. Advancements in our understanding of inborn errors of metabolism are leading to an increasing number of therapeutic options. Many of these treatments can improve long-term neurodevelopment and seizure control. However, the treatments are frequently condition-specific. A high index of suspicion is required for prompt identification and treatment. When suspected, simultaneous metabolic and molecular testing are recommended along with concurrent treatment.
Collapse
Affiliation(s)
- William Burns
- Division of Genetics and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH.
| | - Bimal P Chaudhari
- Division of Genetics and Genomic Medicine, Nationwide Children's Hospital, Columbus, OH; Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH; Division of Neonatology, Nationwide Children's Hospital, Columbus, OH; Steve and Cindy Rasmussen Institute for Genomic Medicine, Nationwide Children's Hospital, Columbus, OH
| | - Darrah N Haffner
- Department of Pediatrics, The Ohio State University College of Medicine, Columbus, OH; Division of Neurology, Nationwide Children's Hospital, Columbus, OH
| |
Collapse
|
27
|
Khoja S, Lambert J, Nitzahn M, Eliav A, Zhang Y, Tamboline M, Le CT, Nasser E, Li Y, Patel P, Zhuravka I, Lueptow LM, Tkachyova I, Xu S, Nissim I, Schulze A, Lipshutz GS. Gene therapy for guanidinoacetate methyltransferase deficiency restores cerebral and myocardial creatine while resolving behavioral abnormalities. Mol Ther Methods Clin Dev 2022; 25:278-296. [PMID: 35505663 PMCID: PMC9051621 DOI: 10.1016/j.omtm.2022.03.015] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Accepted: 03/27/2022] [Indexed: 11/06/2022]
Abstract
Creatine deficiency disorders are inborn errors of creatine metabolism, an energy homeostasis molecule. One of these, guanidinoacetate N-methyltransferase (GAMT) deficiency, has clinical characteristics that include features of autism, self-mutilation, intellectual disability, and seizures, with approximately 40% having a disorder of movement; failure to thrive can also be a component. Along with low creatine levels, guanidinoacetic acid (GAA) toxicity has been implicated in the pathophysiology of the disorder. Present-day therapy with oral creatine to control GAA lacks efficacy; seizures can persist. Dietary management and pharmacological ornithine treatment are challenging. Using an AAV-based gene therapy approach to express human codon-optimized GAMT in hepatocytes, in situ hybridization, and immunostaining, we demonstrated pan-hepatic GAMT expression. Serial collection of blood demonstrated a marked early and sustained reduction of GAA with normalization of plasma creatine; urinary GAA levels also markedly declined. The terminal time point demonstrated marked improvement in cerebral and myocardial creatine levels. In conjunction with the biochemical findings, treated mice gained weight to nearly match their wild-type littermates, while behavioral studies demonstrated resolution of abnormalities; PET-CT imaging demonstrated improvement in brain metabolism. In conclusion, a gene therapy approach can result in long-term normalization of GAA with increased creatine in guanidinoacetate N-methyltransferase deficiency and at the same time resolves the behavioral phenotype in a murine model of the disorder. These findings have important implications for the development of a new therapy for this abnormality of creatine metabolism.
Collapse
Affiliation(s)
- Suhail Khoja
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Jenna Lambert
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Matthew Nitzahn
- Molecular Biology Institute, UCLA, Los Angeles, CA 90025, USA
| | - Adam Eliav
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - YuChen Zhang
- Semel Institute for Neuroscience, UCLA, Los Angeles, CA 90025, USA
| | - Mikayla Tamboline
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA 90025, USA.,Departments of Molecular and Medical Pharmacology, Universtiy of California, Los Angeles, CA 90025, USA
| | - Colleen T Le
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Eram Nasser
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Yunfeng Li
- Departments of Pathology and Laboratory Medicine, UCLA, Los Angeles, CA 90025, USA
| | - Puja Patel
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA
| | - Irina Zhuravka
- Behavioral Testing Core, Department of Psychology, UCLA, Los Angeles, CA 90025, USA
| | - Lindsay M Lueptow
- Behavioral Testing Core, Department of Psychology, UCLA, Los Angeles, CA 90025, USA
| | - Ilona Tkachyova
- Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Shili Xu
- Crump Institute for Molecular Imaging, UCLA, Los Angeles, CA 90025, USA.,Departments of Molecular and Medical Pharmacology, Universtiy of California, Los Angeles, CA 90025, USA.,Jonsson Comprehensive Cancer Center at UCLA, David Geffen School of Medicine at UCLA, Los Angeles, CA 90025, USA
| | - Itzhak Nissim
- Division of Metabolism and Human Genetics, Children's Hospital of Philadelphia, and the Department of Biochemistry and Biophysics, Perlman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Andreas Schulze
- Department of Paediatrics, University of Toronto, Toronto, ON M5G 1X8, Canada.,Department of Biochemistry, University of Toronto, Toronto, ON M5G 1X8, Canada.,Research Institute, The Hospital for Sick Children, Toronto, ON M5G 1X8, Canada
| | - Gerald S Lipshutz
- Department of Surgery, UCLA, Los Angeles, CA 90025, USA.,Molecular Biology Institute, UCLA, Los Angeles, CA 90025, USA.,Semel Institute for Neuroscience, UCLA, Los Angeles, CA 90025, USA.,Departments of Molecular and Medical Pharmacology, Universtiy of California, Los Angeles, CA 90025, USA.,Intellectual and Developmental Disabilities Research Center, UCLA, Los Angeles, CA 90025, USA
| |
Collapse
|
28
|
Shen M, Yang G, Chen Z, Yang K, Dong H, Yin C, Cheng Y, Zhang C, Gu F, Yang Y, Tian Y. Identification of novel variations in SLC6A8 and GAMT genes causing cerebral creatine deficiency syndrome. Clin Chim Acta 2022; 532:29-36. [PMID: 35588794 DOI: 10.1016/j.cca.2022.05.006] [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: 04/06/2022] [Revised: 04/24/2022] [Accepted: 05/05/2022] [Indexed: 11/03/2022]
Abstract
Cerebral creatine deficiency syndromes (CCDSs) are a group of rare mendelian disorders mainly characterized by intellectual disability, movement anomaly, behavior disorder and seizures. SLC6A8, GAMT, and GATM are known genes responsible for CCDS. In this study, seven pediatric patients with developmental delay were recruited and submitted to a series of clinical evaluation, laboratory testing, and genetic analysis. The clinical manifestations and core biochemical indications of each child were basically consistent with the diagnosis of CCDS. Genetic diagnosis determined that all patients were positive for SLC6A8 or GAMT variation. A total of 12 variants were identified in this cohort, including six novel ones. The frequency of these variants, the Revel scores and the conservatism of the affected amino acids support their pathogenicity. Our findings expanded the mutation spectrum of CCDS disorders, and provided solid evidence for the counseling to affected families.
Collapse
Affiliation(s)
- Ming Shen
- Research Center for Translational Medicine Laboratory, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Guangming Yang
- Research Center for Translational Medicine Laboratory, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Zhehui Chen
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Kai Yang
- Prenatal Diagnosis Center, Beijing Obstetrics and Gynecology Hospital, Capital Medical University, Beijing, China
| | - Hui Dong
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Chengliang Yin
- Medical Big Data Research Center, Medical Innovation Research Division of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Yuxuan Cheng
- Birth Defects Prevention and Control Technology Research Center, Medical Research and Innovation Department, Chinese PLA General Hospital, Beijing, China
| | - Chunyan Zhang
- Birth Defects Prevention and Control Technology Research Center, Medical Research and Innovation Department, Chinese PLA General Hospital, Beijing, China
| | - Fangyan Gu
- Clinical Biobank Center, Medical Innovation Research Division of Chinese PLA General Hospital, Beijing, China
| | - Yanling Yang
- Department of Pediatrics, Peking University First Hospital, Beijing, China
| | - Yaping Tian
- Birth Defects Prevention and Control Technology Research Center, Medical Research and Innovation Department, Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
29
|
Perry LD, Hogg SL, Bowdin S, Ambegaonkar G, Parker AP. Fifteen-minute consultation: The efficient investigation of infantile and childhood epileptic encephalopathies in the era of modern genomics. Arch Dis Child Educ Pract Ed 2022; 107:80-87. [PMID: 33414255 DOI: 10.1136/archdischild-2020-320606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 11/05/2020] [Accepted: 12/09/2020] [Indexed: 11/03/2022]
Abstract
The investigation of children presenting with infantile and childhood epileptic encephalopathies (ICEE) is challenging due to diverse aetiologies, overlapping phenotypes and the relatively low diagnostic yield of MRI, electroencephalography (EEG) and biochemical investigations. Careful history and thorough examination remain essential as these may identify an acquired cause or indicate more targeted investigation for a genetic disorder. Whole exome sequencing (WES) with analysis of a panel of candidate epilepsy genes has increased the diagnostic yield. Whole genome sequencing (WGS), particularly as a trio with both parents' DNA, is likely to supersede WES. Modern genomic investigation impacts on the timing and necessity of other testing. We propose a structured approach for children presenting with ICEE where there is diagnostic uncertainty, emphasising the importance of WGS or, if unavailable, WES early in the investigative process. We note the importance of expert review of all investigations, including radiology, neurophysiology and biochemistry, to confirm the technique used was appropriate as well as the results. It is essential to counsel families on the risks associated with the procedures, the yield of the procedures, findings that are difficult to interpret and implication of 'negative' results. Where children remain without a diagnosis despite comprehensive investigation, we note the importance of ongoing multidisciplinary care.
Collapse
Affiliation(s)
- Luke Daniel Perry
- Developmental Neurosciences, University College London, Great Ormond Street Institute of Child Health, London, UK
| | - Sarah Louise Hogg
- Biochemical Genetics Unit, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Sarah Bowdin
- Department of Clinical Genetics, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Gautam Ambegaonkar
- Paediatric Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| | - Alasdair Pj Parker
- Paediatric Neurology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, UK
| |
Collapse
|
30
|
Vasquez A, Buraniqi E, Wirrell EC. New and emerging pharmacologic treatments for developmental and epileptic encephalopathies. Curr Opin Neurol 2022; 35:145-154. [PMID: 35102126 DOI: 10.1097/wco.0000000000001029] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
PURPOSE OF REVIEW Summarize evidence on Developmental and Epileptic Encephalopathies (DEEs) treatments focusing on new and emerging pharmacologic therapies (see Video, http://links.lww.com/CONR/A61, Supplementary Digital Content 1, which provides an overview of the review). RECENT FINDINGS Advances in the fields of molecular genetics and neurobiology have led to the recognition of underlying pathophysiologic mechanisms involved in an increasing number of DEEs that could be targeted with precision therapies or repurposed drugs, some of which are currently being evaluated in clinical trials. Prompt, optimal therapy is critical, and promising therapies approved or in clinical trials for tuberous sclerosis complex, Dravet and Lennox-Gastaut Syndromes including mammalian target of rapamycin inhibitors, selective membrane channel and antisense oligonucleotide modulation, and repurposed drugs such as fenfluramine, stiripentol and cannabidiol, among others, may improve seizure burden and neurological outcomes. There is an urgent need for collaborative efforts to evaluate the efficacy and safety of emerging DEEs therapies. SUMMARY Development of new therapies promise to address unmet needs for patients with DEEs, including improvement of neurocognitive function and quality of life.
Collapse
Affiliation(s)
- Alejandra Vasquez
- Division of Child and Adolescent Neurology, Department of Neurology, Mayo Clinic, Rochester, Minnesota, USA
| | | | | |
Collapse
|
31
|
Passi GR, Pandey S, Devi ARR, Konanki R, Jain AR, Bhatnagar S, Tripathi R, Jain V. Cerebral creatine deficiency disorders - A clinical, genetic and follow up study from India. Brain Dev 2022; 44:271-280. [PMID: 34974949 DOI: 10.1016/j.braindev.2021.12.004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Revised: 12/04/2021] [Accepted: 12/14/2021] [Indexed: 11/28/2022]
Abstract
INTRODUCTION Cerebral creatine deficiency syndromes (CCDS) are a group of potentially treatable neurometabolic disorders. The clinical, genetic profile and follow up outcome of Indian CCDS patients is presented. MATERIALS AND METHODS This was a retrospective cohort of CCDS patients seen over six-years. Diagnosis was based either on low creatine peak on proton magnetic resonance spectroscopy (MRS) and/or genetic evaluation. RESULTS Thirteen patients were eligible [8 creatine transporter deficiency (CTD), 4 guanidinoacetate methyltransferase (GAMT) deficiency and 1 could not be classified]. The mean (±SD) age at diagnosis was 7.2(±5.0) years. Clinical manifestations included intellectual disability (ID) with significant expressive speech delay in all. Most had significant behavior issues (8/13) and/or autism (8/13). All had history of convulsive seizures (11/13 had epilepsy; 2 patients only had febrile seizures) and 2/13 had movement disorder. Constipation was the commonest non-neurological manifestation (5/13 patients). Cranial MRI was normal in all CTD patients but showed globus pallidus hyperintensity in all four with GAMT deficiency. MRS performed in 11/13 patients, revealed abnormally low creatine peak. A causative genetic variant (novel mutation in nine) was identified in 12 patients. Three GAMT deficiency and one CTD patient reported neurodevelopmental improvement and good seizure control after creatine supplementation. CONCLUSION Intellectual disability, disproportionate speech delay, autism, and epilepsy, were common in our CCDS patients. A normal structural neuroimaging with easily controlled febrile and/or afebrile seizures differentiated CTD from GAMT deficiency patients who had abnormal neuroimaging and often difficult to control epilepsy and movement disorder.
Collapse
Affiliation(s)
- Gouri Rao Passi
- Department of Pediatrics & Pediatric Neurology, Choithram Hospital & Research Centre, Indore, India
| | - Swati Pandey
- Department of Pediatrics & Pediatric Neurology, Santokba Durlabhji Memorial Hospital, Jaipur, India
| | - Akella Radha Rama Devi
- Department of Genetics and Pediatric Neurology, Rainbow Children's Hospital, Hyderabad, India
| | - Ramesh Konanki
- Department of Genetics and Pediatric Neurology, Rainbow Children's Hospital, Hyderabad, India
| | - Abhishek Ravindra Jain
- Department of Genetics and Pediatric Neurology, Rainbow Children's Hospital, Hyderabad, India
| | - Shweta Bhatnagar
- Department of Radio Diagnosis, Sri Aurobindo Institute of Medical Sciences, Indore, India
| | - Ruchi Tripathi
- Department of Pediatrics & Pediatric Neurology, Choithram Hospital & Research Centre, Indore, India
| | - Vivek Jain
- Department of Pediatrics & Pediatric Neurology, Santokba Durlabhji Memorial Hospital, Jaipur, India.
| |
Collapse
|
32
|
Jomura R, Akanuma SI, Tachikawa M, Hosoya KI. SLC6A and SLC16A family of transporters: Contribution to transport of creatine and creatine precursors in creatine biosynthesis and distribution. BIOCHIMICA ET BIOPHYSICA ACTA. BIOMEMBRANES 2022; 1864:183840. [PMID: 34921896 DOI: 10.1016/j.bbamem.2021.183840] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 12/06/2021] [Accepted: 12/09/2021] [Indexed: 11/16/2022]
Abstract
Creatine (Cr) is needed to maintain high energy levels in cells. Since Cr plays reportedly a critical role in neurodevelopment and the immune system, Cr dynamics should be strictly regulated to control these physiological events. This review focuses on the role of transporters that recognize Cr and/or Cr precursors. Our previous studies revealed physiological roles of SLC6A and SLC16A family transporters in Cr dynamics. Creatine transporter (CRT/SLC6A8) contributes to the influx transport of Cr in Cr distribution. γ-Aminobutyric acid transporter 2 (GAT2/SLC6A13) mediates incorporation of guanidinoacetate (GAA), a Cr precursor, in the process of Cr biosynthesis. Monocarboxylate transporter 12 (MCT12/SLC16A12) functions as an efflux transporter for Cr and GAA, and contributes to the process of Cr biosynthesis. Accordingly, the SLC6A and SLC16A family of transporters play important roles in the process of Cr biosynthesis and distribution via permeation of Cr and Cr precursors across the plasma membrane.
Collapse
Affiliation(s)
- Ryuta Jomura
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Shin-Ichi Akanuma
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| | - Masanori Tachikawa
- Graduate School of Biomedical Sciences, Tokushima University, 1-78-1 Shomachi, Tokushima 770-8505, Japan.
| | - Ken-Ichi Hosoya
- Department of Pharmaceutics, Graduate School of Medicine and Pharmaceutical Sciences, University of Toyama, 2630 Sugitani, Toyama 930-0194, Japan.
| |
Collapse
|
33
|
Wojcik M, Morrissey M, Borden K, Teta B, Sicko R, Showers A, Sunny S, Caggana M. Method modification to reduce false positives for newborn screening of guanidinoacetate methyltransferase deficiency. Mol Genet Metab 2022; 135:186-192. [PMID: 35120844 DOI: 10.1016/j.ymgme.2022.01.101] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 01/21/2022] [Accepted: 01/22/2022] [Indexed: 10/19/2022]
Abstract
Guanidinoacetate methyltransferase (GAMT) deficiency is an autosomal recessive disorder that results in reduced activity of guanidinoacetate methyltransferase, an accumulation of guanidinoacetate (GUAC), and a lack of cerebral creatine (CRE). Lack of CRE in the brain can cause intellectual disability, autistic-like behavior, seizures, and movement disorders. Identification at birth and immediate therapy can prevent intellectual disability and seizures. If started early in life, treatment with creatine supplements is highly effective. Because there are reliable biomarkers for GAMT deficiency, GUAC and CRE, and because the disorder is readily treatable with a significant improvement in outcomes, GAMT deficiency is an excellent candidate for newborn screening. Several programs have conducted pilot programs or started screening. An isobaric interferant of the GUAC marker has been reported which may cause false positive results. To reduce the number of false positives, a second-tier HPLC test to separate GUAC from unknown, isobaric interferants may be incorporated into the screening algorithm. New York State began screening for GAMT deficiency in October 2018 using a three-tiered screening approach. Quantification of GUAC and CRE were incorporated into routine screening for amino acids and acylcarnitines. In the first year of screening a total of 263,739 samples were tested for GAMT deficiency. Of these, 3382 required second tier testing. After second tier testing, 210 repeat specimens were requested for borderline results and 10 referrals were made to specialty care centers for confirmatory testing. In the first year of screening there were no confirmed cases of GAMT deficiency detected. To reduce the number of samples needing second tier testing and the number false positives we explored the use of a second MS transition to confirm the identity of the GUAC marker. GUAC and its internal standard are detected as butylated esters after sample preparation and derivatization. The original method used transition of the GUAC molecular ion of m/z 174.1 to a reactant ion of m/z 101.1. To confirm the identity of the GUAC marker we selected a qualifier ion of 174.1 > 73. The alternative product ion results were found to agree more closely with the second tier HPLC-MS/MS results for GUAC. It was found that the alternative transition may be used for quantification of the GUAC marker with acceptable analytical performance (linearity, accuracy, and precision). On March 5, 2020, the method of analysis for GUAC was modified to use the alternative product ion. For a comparable 6-month period, the modified method reduced the number of samples requiring second tier testing by 98%, reduced the number of borderline results requiring a repeat sample by 87.5%, and reduced the number of referrals to specialty care centers by 85%. Using the modified method, the correlation (r-squared) of the first and second tier screening results for GUAC is greater than 0.95. Since the first-tier results correlate well with the second-tier results, the second-tier screening is no longer necessary with the modified method.
Collapse
Affiliation(s)
- Matthew Wojcik
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Mark Morrissey
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA.
| | - Kimberly Borden
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Bianca Teta
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Robert Sicko
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Amanda Showers
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Sherly Sunny
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Michele Caggana
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| |
Collapse
|
34
|
Whitehead MT, Lai LM, Blüml S. Clinical 1H MRS in childhood neurometabolic diseases — part 2: MRS signatures. Neuroradiology 2022; 64:1111-1126. [DOI: 10.1007/s00234-022-02918-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Accepted: 02/10/2022] [Indexed: 12/23/2022]
|
35
|
McGinn RJ, Von Stein EL, Summers Stromberg JE, Li Y. Precision medicine in epilepsy. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2022; 190:147-188. [DOI: 10.1016/bs.pmbts.2022.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Fernandes-Pires G, Braissant O. Current and potential new treatment strategies for creatine deficiency syndromes. Mol Genet Metab 2022; 135:15-26. [PMID: 34972654 DOI: 10.1016/j.ymgme.2021.12.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2021] [Revised: 12/14/2021] [Accepted: 12/14/2021] [Indexed: 12/16/2022]
Abstract
Creatine deficiency syndromes (CDS) are inherited metabolic disorders caused by mutations in GATM, GAMT and SLC6A8 and mainly affect central nervous system (CNS). AGAT- and GAMT-deficient patients lack the functional brain endogenous creatine (Cr) synthesis pathway but express the Cr transporter SLC6A8 at blood-brain barrier (BBB), and can thus be treated by oral supplementation of high doses of Cr. For Cr transporter deficiency (SLC6A8 deficiency or CTD), current treatment strategies benefit one-third of patients. However, as their phenotype is not completely reversed, and for the other two-thirds of CTD patients, the development of novel more effective therapies is needed. This article aims to review the current knowledge on Cr metabolism and CDS clinical aspects, highlighting their current treatment possibilities and the most recent research perspectives on CDS potential therapeutics designed, in particular, to bring new options for the treatment of CTD.
Collapse
Affiliation(s)
- Gabriella Fernandes-Pires
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland
| | - Olivier Braissant
- Service of Clinical Chemistry, University of Lausanne and Lausanne University Hospital, Lausanne, Switzerland.
| |
Collapse
|
37
|
Whitehead MT, Bluml S. Proton and Multinuclear Spectroscopy of the Pediatric Brain. Magn Reson Imaging Clin N Am 2021; 29:543-555. [PMID: 34717844 DOI: 10.1016/j.mric.2021.06.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Magnetic resonance spectroscopy (MRS) is a valuable adjunct to structural brain imaging. State-of-the-art MRS has benefited greatly from recent technical advancements. Neurometabolic alterations in pediatric brain diseases have implications for diagnosis, prognosis, and therapy. Herein, the authors discuss MRS technical considerations and applications in the setting of various pediatric disease processes including tumors, metabolic diseases, hypoxic/ischemic encephalopathy/stroke, epilepsy, demyelinating disease, and infection.
Collapse
Affiliation(s)
- Matthew T Whitehead
- Department of Radiology, Children's National Hospital, 111 Michigan Avenue NW, Washington, DC 20010, USA; Prenatal Pediatrics Institute, Children's National Hospital, Washington, DC, USA; The George Washington University School of Medicine and Health Sciences, Washington, DC, USA.
| | - Stefan Bluml
- Department of Radiology, Children's Hospital Los Angeles, Keck School of Medicine, University of Southern California, 450 Sunset Boulevard, Los Angeles, CA 90027, USA; Rudi Schulte Research Institute, Santa Barbara, CA, USA
| |
Collapse
|
38
|
Balestrini S, Chiarello D, Gogou M, Silvennoinen K, Puvirajasinghe C, Jones WD, Reif P, Klein KM, Rosenow F, Weber YG, Lerche H, Schubert-Bast S, Borggraefe I, Coppola A, Troisi S, Møller RS, Riva A, Striano P, Zara F, Hemingway C, Marini C, Rosati A, Mei D, Montomoli M, Guerrini R, Cross JH, Sisodiya SM. Real-life survey of pitfalls and successes of precision medicine in genetic epilepsies. J Neurol Neurosurg Psychiatry 2021; 92:1044-1052. [PMID: 33903184 PMCID: PMC8458055 DOI: 10.1136/jnnp-2020-325932] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2020] [Revised: 02/20/2021] [Accepted: 03/28/2021] [Indexed: 01/05/2023]
Abstract
OBJECTIVE The term 'precision medicine' describes a rational treatment strategy tailored to one person that reverses or modifies the disease pathophysiology. In epilepsy, single case and small cohort reports document nascent precision medicine strategies in specific genetic epilepsies. The aim of this multicentre observational study was to investigate the deeper complexity of precision medicine in epilepsy. METHODS A systematic survey of patients with epilepsy with a molecular genetic diagnosis was conducted in six tertiary epilepsy centres including children and adults. A standardised questionnaire was used for data collection, including genetic findings and impact on clinical and therapeutic management. RESULTS We included 293 patients with genetic epilepsies, 137 children and 156 adults, 162 females and 131 males. Treatment changes were undertaken because of the genetic findings in 94 patients (32%), including rational precision medicine treatment and/or a treatment change prompted by the genetic diagnosis, but not directly related to known pathophysiological mechanisms. There was a rational precision medicine treatment for 56 patients (19%), and this was tried in 33/56 (59%) and was successful (ie, >50% seizure reduction) in 10/33 (30%) patients. In 73/293 (25%) patients there was a treatment change prompted by the genetic diagnosis, but not directly related to known pathophysiological mechanisms, and this was successful in 24/73 (33%). SIGNIFICANCE Our survey of clinical practice in specialised epilepsy centres shows high variability of clinical outcomes following the identification of a genetic cause for an epilepsy. Meaningful change in the treatment paradigm after genetic testing is not yet possible for many people with epilepsy. This systematic survey provides an overview of the current application of precision medicine in the epilepsies, and suggests the adoption of a more considered approach.
Collapse
Affiliation(s)
- Simona Balestrini
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, and Chalfont Centre for Epilepsy, Gerrard Cross, UK
- Neurology Unit and Neurogenetics Laboratories, Meyer Children Hospital, Florence, Italy
| | - Daniela Chiarello
- Institute of Child Health, University College of London (UCL) Great Ormond Street NIHR BRC, London, UK
- Great Ormond Street Hospital for Children, London, UK
| | - Maria Gogou
- Institute of Child Health, University College of London (UCL) Great Ormond Street NIHR BRC, London, UK
- Great Ormond Street Hospital for Children, London, UK
| | - Katri Silvennoinen
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, and Chalfont Centre for Epilepsy, Gerrard Cross, UK
| | | | - Wendy D Jones
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, and Chalfont Centre for Epilepsy, Gerrard Cross, UK
- Institute of Child Health, University College of London (UCL) Great Ormond Street NIHR BRC, London, UK
- Great Ormond Street Hospital for Children, London, UK
| | - Philipp Reif
- Epilepsy Center Frankfurt Rhine-Main University of Frankfurt, University of Frankfurt, Frankfurt Rhine Main, Germany
- Department of Neurology, University Hospital Frankfurt and LOEWE Center for Personalized Translational Epilepsy Research (CePTER) Goethe-University Frankfurt, Frankfurt am Main, Germany
- Epilepsy Center Hessen and Department of Neurology, Philipps-University, Marburg, Germany
| | - Karl Martin Klein
- Epilepsy Center Frankfurt Rhine-Main University of Frankfurt, University of Frankfurt, Frankfurt Rhine Main, Germany
- Department of Neurology, University Hospital Frankfurt and LOEWE Center for Personalized Translational Epilepsy Research (CePTER) Goethe-University Frankfurt, Frankfurt am Main, Germany
- Epilepsy Center Hessen and Department of Neurology, Philipps-University, Marburg, Germany
- Departments of Clinical Neurosciences, Medical Genetics and Community Health Sciences, Hotchkiss Brain Institute & Alberta Children's Hospital Research Institute, Cumming School of Medicine, University of Calgary, Calgary, Alberta, Canada
| | - Felix Rosenow
- Epilepsy Center Frankfurt Rhine-Main University of Frankfurt, University of Frankfurt, Frankfurt Rhine Main, Germany
- Department of Neurology, University Hospital Frankfurt and LOEWE Center for Personalized Translational Epilepsy Research (CePTER) Goethe-University Frankfurt, Frankfurt am Main, Germany
- Epilepsy Center Hessen and Department of Neurology, Philipps-University, Marburg, Germany
| | - Yvonne G Weber
- Department of Neurology and Epileptology, University of Tübingen, Tubingen, Germany
- Department of Epileptology and Neurology, University of Aachen, Aachen, Germany
| | - Holger Lerche
- Epilepsy Center Frankfurt Rhine-Main University of Frankfurt, University of Frankfurt, Frankfurt Rhine Main, Germany
- Department of Neurology and Epileptology, University of Tübingen, Tubingen, Germany
| | - Susanne Schubert-Bast
- Epilepsy Center Frankfurt Rhine-Main University of Frankfurt, University of Frankfurt, Frankfurt Rhine Main, Germany
| | - Ingo Borggraefe
- Department of Pediatric Neurology, Dr von Haunerschen Kinderspital, University of Munich, Munich, Germany
| | - Antonietta Coppola
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Epilepsy Centre, Federico II University, Naples, Italy
| | - Serena Troisi
- Department of Neuroscience, Reproductive and Odontostomatological Sciences, Epilepsy Centre, Federico II University, Naples, Italy
| | - Rikke S Møller
- The Danish Epilepsy Centre Filadelfia, Dianalund, and Institute for Regional Health Services Research, University of Southern Denmark, Odense, Denmark
| | - Antonella Riva
- Department of Neurosciences, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
| | - Pasquale Striano
- Department of Neurosciences, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
- IRCCS 'G. Gaslini' Institute, Genova, Italy
| | - Federico Zara
- Department of Neurosciences, Rehabilitation, Ophtalmology, Genetics, Maternal and Child Health, University of Genova, Genova, Italy
- IRCCS 'G. Gaslini' Institute, Genova, Italy
| | | | - Carla Marini
- Neurology Unit and Neurogenetics Laboratories, Meyer Children Hospital, Florence, Italy
- Child Neurology and Psychiatric Unit, Salesi Children's Hospital, Ancona, Italy
| | - Anna Rosati
- Neurology Unit and Neurogenetics Laboratories, Meyer Children Hospital, Florence, Italy
| | - Davide Mei
- Neurology Unit and Neurogenetics Laboratories, Meyer Children Hospital, Florence, Italy
| | - Martino Montomoli
- Neurology Unit and Neurogenetics Laboratories, Meyer Children Hospital, Florence, Italy
| | - Renzo Guerrini
- Neurology Unit and Neurogenetics Laboratories, Meyer Children Hospital, Florence, Italy
| | - J Helen Cross
- Institute of Child Health, University College of London (UCL) Great Ormond Street NIHR BRC, London, UK
- Great Ormond Street Hospital for Children, London, UK
| | - Sanjay M Sisodiya
- Department of Clinical and Experimental Epilepsy, UCL Queen Square Institute of Neurology, London, and Chalfont Centre for Epilepsy, Gerrard Cross, UK
| |
Collapse
|
39
|
Hart K, Rohrwasser A, Wallis H, Golsan H, Shao J, Anderson T, Wang X, Szabo-Fresnais N, Morrissey M, Kay DM, Wojcik M, Galvin-Parton PA, Longo N, Caggana M, Pasquali M. Prospective identification by neonatal screening of patients with guanidinoacetate methyltransferase deficiency. Mol Genet Metab 2021; 134:60-64. [PMID: 34389248 DOI: 10.1016/j.ymgme.2021.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 11/17/2022]
Abstract
INTRODUCTION Guanidinoacetate methyltransferase (GAMT) deficiency is an inherited metabolic disorder that impairs the synthesis of creatine (CRE). Lack of CRE in the brain can cause intellectual disability, autistic-like behavior, seizures, and movement disorders. Identification at birth and immediate therapy can prevent intellectual disability and seizures. Here we report the first two cases of GAMT deficiency identified at birth by newborn screening (NBS) in Utah and New York. METHODS NBS dried blood spots were analyzed by tandem mass spectrometry (MS/MS) using either derivatized or non-derivatized assays to detect guanidinoacetate (GUAC) and CRE. For any positive samples, a second-tier test using a more selective method, ultra-performance liquid chromatography (UPLC) combined with MS/MS, was performed to separate GUAC from potential isobaric interferences. RESULTS NBS for GAMT deficiency began in Utah on June 1, 2015 using a derivatized method for the detection of GUAC and CRE. In May 2019, the laboratory and method transitioned to a non-derivatized method. GAMT screening was added to the New York State NBS panel on October 1, 2018 using a derivatized method. In New York, a total of 537,408 babies were screened, 23 infants were referred and one newborn was identified with GAMT deficiency. In Utah, a total of 273,902 infants were screened (195,425 with the derivatized method, 78,477 with the non-derivatized method), three infants referred and one was identified with GAMT deficiency. Mean levels of GUAC and CRE were similar between methods (Utah derivatized: GUAC = 1.20 ± 0.43 μmol/L, CRE = 238 ± 96 μmol/L; Utah non-derivatized: GUAC = 1.23 ± 0.61 μmol/L, CRE = 344 ± 150 μmol/L, New York derivatized: GUAC = 1.34 ± 0.57 μmol/L, CRE = 569 ± 155 μmol/L). With either Utah method, similar concentrations of GUAC are observed in first (collected around 1 day of age) and the second NBS specimens (routinely collected at 7-16 days of age), while CRE concentrations decreased in the second NBS specimens. Both infants identified with GAMT deficiency started therapy by 2 weeks of age and are growing and developing normally at 7 (Utah) and 4 (New York) months of age. CONCLUSIONS Newborn screening allows for the prospective identification of GAMT deficiency utilizing elevated GUAC concentration as a marker. First-tier screening may be incorporated into existing methods for amino acids and acylcarnitines without the need for new equipment or staff. Newborn screening performed by either derivatized or non-derivatized methods and coupled with second-tier testing, has a very low false positive rate and can prospectively identify affected children. SummaryCerebral creatine deficiency syndromes caused by defects in creatine synthesis can result in intellectual disability, and are preventable if therapy is initiated early in life. This manuscript reports the identification of two infants with GAMT deficiency (one of the cerebral creatine deficiency syndromes) by newborn screening and demonstrates NBS feasibility using a variety of methods.
Collapse
Affiliation(s)
- Kim Hart
- Utah Department of Health, Salt Lake City, UT, USA.
| | | | - Heidi Wallis
- Utah Department of Health, Salt Lake City, UT, USA; Association for Creatine Deficiencies, Carlsbad, CA, USA
| | | | - Jianyin Shao
- Utah Department of Health, Salt Lake City, UT, USA
| | | | - Xiaoli Wang
- Utah Department of Health, Salt Lake City, UT, USA
| | | | - Mark Morrissey
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Denise M Kay
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Matthew Wojcik
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | | | - Nicola Longo
- Department of Pathology, University of Utah, ARUP Laboratories, Salt Lake City, UT, USA; Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA
| | - Michele Caggana
- Newborn Screening Program, Wadsworth Center, New York State Department of Health, Albany, NY, USA
| | - Marzia Pasquali
- Department of Pathology, University of Utah, ARUP Laboratories, Salt Lake City, UT, USA; Division of Medical Genetics, Department of Pediatrics, University of Utah, Salt Lake City, UT, USA.
| |
Collapse
|
40
|
Intellectual Disability and Brain Creatine Deficit: Phenotyping of the Genetic Mouse Model for GAMT Deficiency. Genes (Basel) 2021; 12:genes12081201. [PMID: 34440375 PMCID: PMC8391262 DOI: 10.3390/genes12081201] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 07/23/2021] [Accepted: 07/25/2021] [Indexed: 11/17/2022] Open
Abstract
Guanidinoacetate methyltransferase deficiency (GAMT-D) is one of three cerebral creatine (Cr) deficiency syndromes due to pathogenic variants in the GAMT gene (19p13.3). GAMT-D is characterized by the accumulation of guanidinoacetic acid (GAA) and the depletion of Cr, which result in severe global developmental delay (and intellectual disability), movement disorder, and epilepsy. The GAMT knockout (KO) mouse model presents biochemical alterations in bodily fluids, the brain, and muscles, including increased GAA and decreased Cr and creatinine (Crn) levels, which are similar to those observed in humans. At the behavioral level, only limited and mild alterations have been reported, with a large part of analyzed behaviors being unaffected in GAMT KO as compared with wild-type mice. At the cerebral level, decreased Cr and Crn and increased GAA and other guanidine compound levels have been observed. Nevertheless, the effects of Cr deficiency and GAA accumulation on many neurochemical, morphological, and molecular processes have not yet been explored. In this review, we summarize data regarding behavioral and cerebral GAMT KO phenotypes, and focus on uncharted behavioral alterations that are comparable with the clinical symptoms reported in GAMT-D patients, including intellectual disability, poor speech, and autistic-like behaviors, as well as unexplored Cr-induced cerebral alterations.
Collapse
|
41
|
Soo AKS, Ferrini A, Kurian MA. Precision medicine for genetic childhood movement disorders. Dev Med Child Neurol 2021; 63:925-933. [PMID: 33763868 DOI: 10.1111/dmcn.14869] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 02/23/2021] [Indexed: 12/22/2022]
Abstract
Increasingly effective targeted precision medicine is either already available or in development for a number of genetic childhood movement disorders. Patient-centred, personalized approaches include the repurposing of existing treatments for specific conditions and the development of novel therapies that target the underlying genetic defect or disease mechanism. In tandem with these scientific advances, close collaboration between clinicians, researchers, affected families, and stakeholders in the wider community will be key to successfully delivering such precision therapies to children with movement disorders. What this paper adds Precision medicine for genetic childhood movement disorders is developing rapidly. Accurate diagnosis, disease-specific outcome measures, and collaborative multidisciplinary work will accelerate the progress of such strategies.
Collapse
Affiliation(s)
- Audrey K S Soo
- Developmental Neurosciences, UCL Great Ormond Street Hospital, Zayed Centre for Research into Rare Disease in Children, London, UK.,Department of Paediatric Neurology, Great Ormond Street Hospital, London, UK
| | - Arianna Ferrini
- Developmental Neurosciences, UCL Great Ormond Street Hospital, Zayed Centre for Research into Rare Disease in Children, London, UK
| | - Manju A Kurian
- Developmental Neurosciences, UCL Great Ormond Street Hospital, Zayed Centre for Research into Rare Disease in Children, London, UK.,Department of Paediatric Neurology, Great Ormond Street Hospital, London, UK
| |
Collapse
|
42
|
Almannai M, Al Mahmoud RA, Mekki M, El-Hattab AW. Metabolic Seizures. Front Neurol 2021; 12:640371. [PMID: 34295297 PMCID: PMC8290068 DOI: 10.3389/fneur.2021.640371] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2020] [Accepted: 05/21/2021] [Indexed: 12/13/2022] Open
Abstract
Metabolic diseases should always be considered when evaluating children presenting with seizures. This is because many metabolic disorders are potentially treatable and seizure control can be achieved when these diseases are appropriately treated. Seizures caused by underlying metabolic diseases (metabolic seizures) should be particularly considered in unexplained neonatal seizures, refractory seizures, seizures related to fasting or food intake, seizures associated with other systemic or neurologic features, parental consanguinity, and family history of epilepsy. Metabolic seizures can be caused by various amino acids metabolic disorders, disorders of energy metabolism, cofactor-related metabolic diseases, purine and pyrimidine metabolic diseases, congenital disorders of glycosylation, and lysosomal and peroxisomal disorders. Diagnosing metabolic seizures without delay is essential because the immediate initiation of appropriate therapy for many metabolic diseases can prevent or minimize complications.
Collapse
Affiliation(s)
- Mohammed Almannai
- Section of Medical Genetics, Children's Hospital, King Fahad Medical City, Riyadh, Saudi Arabia
| | - Rabah A Al Mahmoud
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Pediatrics, University Hospital Sharjah, Sharjah, United Arab Emirates
| | - Mohammed Mekki
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Department of Pediatrics, Al Qassimi Women's and Children's Hospital, Sharjah, United Arab Emirates
| | - Ayman W El-Hattab
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates.,Clinical Genetics, University Hospital Sharjah, Sharjah, United Arab Emirates
| |
Collapse
|
43
|
Modi BP, Khan HN, van der Lee R, Wasim M, Haaxma CA, Richmond PA, Drögemöller B, Shah S, Salomons G, van der Kloet FM, Vaz FM, van der Crabben SN, Ross CJ, Wasserman WW, van Karnebeek CD, Awan FR. Adult GAMT deficiency: A literature review and report of two siblings. Mol Genet Metab Rep 2021; 27:100761. [PMID: 33996490 PMCID: PMC8093930 DOI: 10.1016/j.ymgmr.2021.100761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Accepted: 04/18/2021] [Indexed: 11/02/2022] Open
Abstract
Guanidinoacetate methyltransferase (GAMT) deficiency is a creatine deficiency disorder and an inborn error of metabolism presenting with progressive intellectual and neurological deterioration. As most cases are identified and treated in early childhood, adult phenotypes that can help in understanding the natural history of the disorder are rare. We describe two adult cases of GAMT deficiency from a consanguineous family in Pakistan that presented with a history of global developmental delay, cognitive impairments, excessive drooling, behavioral abnormalities, contractures and apparent bone deformities initially presumed to be the reason for abnormal gait. Exome sequencing identified a homozygous nonsense variant in GAMT: NM_000156.5:c.134G>A (p.Trp45*). We also performed a literature review and compiled the genetic and clinical characteristics of all adult cases of GAMT deficiency reported to date. When compared to the adult cases previously reported, the musculoskeletal phenotype and the rapidly progressive nature of neurological and motor decline seen in our patients is striking. This study presents an opportunity to gain insights into the adult presentation of GAMT deficiency and highlights the need for in-depth evaluation and reporting of clinical features to expand our understanding of the phenotypic spectrum.
Collapse
Affiliation(s)
- Bhavi P. Modi
- Centre for Molecular Medicine and Therapeutics, Dept. of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Correspondence to: B. P. Modi, University of British Columbia, BC Children's Hospital Research Institute, 938 W 28 Ave, Vancouver, BC V5Z 4H4, Canada.
| | - Haq Nawaz Khan
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Robin van der Lee
- Centre for Molecular Medicine and Therapeutics, Dept. of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Muhammad Wasim
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
| | - Charlotte A. Haaxma
- Department of Pediatric Neurology, Amalia Children's Hospital, Radboud University Medical Centre, Nijmegen, the Netherlands
| | - Phillip A. Richmond
- Centre for Molecular Medicine and Therapeutics, Dept. of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Britt Drögemöller
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Suleman Shah
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
| | - Gajja Salomons
- Laboratory for Genetic Metabolic Diseases, Amsterdam University Medical Centres, Amsterdam, the Netherlands
| | - Frans M. van der Kloet
- Laboratory for Genetic Metabolic Diseases, Amsterdam University Medical Centres, Amsterdam, the Netherlands
- Swammerdam Institute for Life Sciences, University of Amsterdam, the Netherlands
| | - Fred M. Vaz
- Laboratory for Genetic Metabolic Diseases, Amsterdam University Medical Centres, Amsterdam, the Netherlands
- Dept. of Clinical Chemistry and Pediatrics, Amsterdam Gastroenterology Endocrinology Metabolism, University of Amsterdam, the Netherlands
| | | | - Colin J. Ross
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC, Canada
| | - Wyeth W. Wasserman
- Centre for Molecular Medicine and Therapeutics, Dept. of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
| | - Clara D.M. van Karnebeek
- Centre for Molecular Medicine and Therapeutics, Dept. of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC, Canada
- Department of Pediatrics, Emma Children's Hospital, Amsterdam University Medical Centres, Amsterdam, Netherlands
- Department of Pediatric Metabolic Diseases, Amalia Children's Hospital, Radboud Centre for Mitochondrial Medicine, Radboud University Medical Centre, Nijmegen, the Netherlands
- United for Metabolic Diseases, the Netherlands
| | - Fazli Rabbi Awan
- Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad, Pakistan
- Pakistan Institute of Engineering and Applied Sciences (PIEAS), Islamabad, Pakistan
- Correspondence to: F. R. Awan, Health Biotechnology Division, National Institute for Biotechnology and Genetic Engineering (NIBGE), Faisalabad 38000, Pakistan.
| |
Collapse
|
44
|
Shi K, Zhao H, Xu S, Han H, Li W. Treatment efficacy of high-dose creatine supplementation in a child with creatine transporter (SLC6A8) deficiency. Mol Genet Genomic Med 2021; 9:e1640. [PMID: 33656256 PMCID: PMC8123749 DOI: 10.1002/mgg3.1640] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2020] [Accepted: 02/10/2021] [Indexed: 01/03/2023] Open
Abstract
Background Creatine transporter deficiency is an inborn error of metabolism caused by a deficiency in the creatine transporter protein encoded by the SLC6A8 gene. Previous treatment with creatine supplementation, either alone or in combination with creatine precursors (arginine or glycine), has been attempted; the efficacy of therapy, however, remains controversial. Methods and Results To analyze the treatment efficacy of high‐dose creatine supplementation on creatine transporter deficiency, we reported a child diagnosed with creatine transporter deficiency, who was treated with a conventional dose of creatine (400 mg/kg/d) for 1 month, then twice the dose (800 mg/kg/d) for 2 months, and finally 3 times the dose (1200 mg/kg/d) for 3 months. The patient tolerated the treatment well and showed improvements in muscle mass and strength when the creatine dose was gradually increased to 1200 mg/kg/d. However, when assessed by proton magnetic resonance spectroscopy (H‐MRS), the brain creatine concentration did not increase, and there was no improvement in speech and neurodevelopmental symptoms. Conclusion We conclude that high‐dose creatine supplementation (1200 mg/kg/d) alone improved muscular symptoms, but did not improve cognitive symptoms and brain creatine concentration assessed using H‐MRS. Therefore, new treatment strategies are required for the management of creatine transporter deficiency. Creatine transporter deficiency is an inborn error of metabolism caused by a deficiency in the creatine transporter protein encoded by the SLC6A8 gene. Previously treatment with creatine supplementation, either alone or in combination with creatine precursors (arginine or glycine), has been attempted; the efficacy of therapy, however, remains controversial. We conclude that high‐dose creatine supplementation (1200 mg/kg/d) alone improved muscular symptoms, but did not improve cognitive symptoms and brain creatine concentration assessed using proton magnetic resonance spectroscopy. Therefore, new treatment strategies are required for the management of creatine transporter deficiency.
Collapse
Affiliation(s)
- Kaili Shi
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| | - Huimin Zhao
- Department of pediatrics, Shan'xi Medical University, Taiyuan, China
| | - Shuming Xu
- Department of Imaging of Shanxi, Children's Hospital, Taiyuan, China
| | - Hong Han
- Department of Neurology of Shanxi, Children's Hospital, Taiyuan, China
| | - Wenjuan Li
- Department of Neurology, Guangzhou Women and Children's Medical Center, Guangzhou, China
| |
Collapse
|
45
|
Creatine Supplementation and Brain Health. Nutrients 2021; 13:nu13020586. [PMID: 33578876 PMCID: PMC7916590 DOI: 10.3390/nu13020586] [Citation(s) in RCA: 81] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 02/03/2021] [Accepted: 02/04/2021] [Indexed: 01/06/2023] Open
Abstract
There is a robust and compelling body of evidence supporting the ergogenic and therapeutic role of creatine supplementation in muscle. Beyond these well-described effects and mechanisms, there is literature to suggest that creatine may also be beneficial to brain health (e.g., cognitive processing, brain function, and recovery from trauma). This is a growing field of research, and the purpose of this short review is to provide an update on the effects of creatine supplementation on brain health in humans. There is a potential for creatine supplementation to improve cognitive processing, especially in conditions characterized by brain creatine deficits, which could be induced by acute stressors (e.g., exercise, sleep deprivation) or chronic, pathologic conditions (e.g., creatine synthesis enzyme deficiencies, mild traumatic brain injury, aging, Alzheimer’s disease, depression). Despite this, the optimal creatine protocol able to increase brain creatine levels is still to be determined. Similarly, supplementation studies concomitantly assessing brain creatine and cognitive function are needed. Collectively, data available are promising and future research in the area is warranted.
Collapse
|
46
|
Kreider RB, Stout JR. Creatine in Health and Disease. Nutrients 2021; 13:nu13020447. [PMID: 33572884 PMCID: PMC7910963 DOI: 10.3390/nu13020447] [Citation(s) in RCA: 64] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 01/22/2021] [Accepted: 01/27/2021] [Indexed: 12/14/2022] Open
Abstract
Although creatine has been mostly studied as an ergogenic aid for exercise, training, and sport, several health and potential therapeutic benefits have been reported. This is because creatine plays a critical role in cellular metabolism, particularly during metabolically stressed states, and limitations in the ability to transport and/or store creatine can impair metabolism. Moreover, increasing availability of creatine in tissue may enhance cellular metabolism and thereby lessen the severity of injury and/or disease conditions, particularly when oxygen availability is compromised. This systematic review assesses the peer-reviewed scientific and medical evidence related to creatine's role in promoting general health as we age and how creatine supplementation has been used as a nutritional strategy to help individuals recover from injury and/or manage chronic disease. Additionally, it provides reasonable conclusions about the role of creatine on health and disease based on current scientific evidence. Based on this analysis, it can be concluded that creatine supplementation has several health and therapeutic benefits throughout the lifespan.
Collapse
Affiliation(s)
- Richard B. Kreider
- Human Clinical Research Facility, Exercise & Sport Nutrition Lab, Department of Health & Kinesiology, Texas A&M University, College Station, TX 77843, USA
- Correspondence:
| | - Jeffery R. Stout
- Physiology of Work and Exercise Response (POWER) Laboratory, Institute of Exercise Physiology and Rehabilitation Science, School of Kinesiology and Physical Therapy, University of Central Florida, 12494 University Blvd., Orlando, FL 32816, USA;
| |
Collapse
|
47
|
Impact of predictive, preventive and precision medicine strategies in epilepsy. Nat Rev Neurol 2020; 16:674-688. [PMID: 33077944 DOI: 10.1038/s41582-020-0409-4] [Citation(s) in RCA: 47] [Impact Index Per Article: 9.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/01/2020] [Indexed: 12/15/2022]
Abstract
Over the last decade, advances in genetics, neuroimaging and EEG have enabled the aetiology of epilepsy to be identified earlier in the disease course than ever before. At the same time, progress in the study of experimental models of epilepsy has provided a better understanding of the mechanisms underlying the condition and has enabled the identification of therapies that target specific aetiologies. We are now witnessing the impact of these advances in our daily clinical practice. Thus, now is the time for a paradigm shift in epilepsy treatment from a reactive attitude, treating patients after the onset of epilepsy and the initiation of seizures, to a proactive attitude that is more broadly integrated into a 'P4 medicine' approach. This P4 approach, which is personalized, predictive, preventive and participatory, puts patients at the centre of their own care and, ultimately, aims to prevent the onset of epilepsy. This aim will be achieved by adapting epilepsy treatments not only to a given syndrome but also to a given patient and moving from the usual anti-seizure treatments to personalized treatments designed to target specific aetiologies. In this Review, we present the current state of this ongoing revolution, emphasizing the impact on clinical practice.
Collapse
|
48
|
Narayan V, Mahay SB, Verma IC, Puri RD. Case Series of Creatine Deficiency Syndrome due to Guanidinoacetate Methyltransferase Deficiency. Ann Indian Acad Neurol 2020; 23:347-351. [PMID: 32606525 PMCID: PMC7313580 DOI: 10.4103/aian.aian_367_18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2018] [Revised: 12/02/2018] [Accepted: 12/06/2018] [Indexed: 12/27/2022] Open
Abstract
Guanidinoacetate methyltransferase (GAMT) deficiency is the second most common defect in the creatine metabolism pathway resulting in cerebral creatine deficiency syndrome (CCDS). We report three patients from two unrelated families, diagnosed with GAMT deficiency on next-generation sequencing. All the probands had happy predisposition as a predominant manifestation in addition to the reported features of global developmental delay, seizures, and microcephaly. This further expands the phenotype of CCDS. The workup for creatine deficiency disorder should be included in the diagnostic algorithm for children with nonsyndromic intellectual disability, especially in those with a happy demeanor. These cases exemplify the utility of magnetic resonance spectroscopy of the brain in the workup of nonsyndromic intellectual disability to diagnose a potentially treatable disorder. In addition, documentation of low serum creatinine may be supportive. Early diagnosis and treatment is essential for better prognosis.
Collapse
Affiliation(s)
- Vinu Narayan
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Sunita Bijarnia Mahay
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Ishwar Chander Verma
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| | - Ratna Dua Puri
- Institute of Medical Genetics and Genomics, Sir Ganga Ram Hospital, New Delhi, India
| |
Collapse
|
49
|
Sinha A, Ahmed S, George C, Tsagaris M, Naufer A, von Both I, Tkachyova I, van Eede M, Henkelman M, Schulze A. Magnetic resonance imaging reveals specific anatomical changes in the brain of Agat- and Gamt-mice attributed to creatine depletion and guanidinoacetate alteration. J Inherit Metab Dis 2020; 43:827-842. [PMID: 31951021 DOI: 10.1002/jimd.12215] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 12/30/2019] [Accepted: 01/09/2020] [Indexed: 11/07/2022]
Abstract
Arginine:glycine amidinotransferase- and guanidinoacetate methyltransferase deficiency are severe neurodevelopmental disorders. It is not known whether mouse models of disease express a neuroanatomical phenotype. High-resolution magnetic resonance imaging (MRI) with advanced image analysis was performed in perfused, fixed mouse brains encapsulated with the skull from male, 10-12 week old Agat -exc and B6J.Cg-Gamt tm1Isb mice (n = 48; n = 8 per genotype, strain). T2-weighted MRI scans were nonlinearly aligned to a 3D atlas of the mouse brain with 62 structures identified. Local differences in brain shape related to genotype were assessed by analysis of deformation fields. Creatine (Cr) and guanidinoacetate (GAA) were measured with high-performance liquid chromatography (HPLC) in brain homogenates (n = 24; n = 4 per genotype, strain) after whole-body perfusion. Cr was decreased in the brain of Agat- and Gamt mutant mice. GAA was decreased in Agat-/- and increased in Gamt-/- . Body weight and brain volume were lower in Agat-/- than in Gamt-/- . The analysis of entire brain structures revealed corpus callosum, internal capsule, fimbria and hypothalamus being different between the genotypes in both strains. Eighteen and fourteen significant peaks (local areas of difference in relative size) were found in Agat- and Gamt mutants, respectively. Comparing Agat-/- with Gamt-/- , we found changes in three brain regions, lateral septum, amygdala, and medulla. Intra-strain differences in four brain structures can be associated with Cr deficiency, while the inter-strain differences in three brain structures of the mutant mice may relate to GAA. Correlating these neuroanatomical findings with gene expression data implies the role of Cr metabolism in the developing brain and the importance of early intervention in patients with Cr deficiency syndromes.
Collapse
Affiliation(s)
- Ankit Sinha
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Sohail Ahmed
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Chris George
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Melina Tsagaris
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Amriya Naufer
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ingo von Both
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Ilona Tkachyova
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Matthijs van Eede
- Mouse Imaging Centre, Toronto Center of Phenogenomics, Toronto, Ontario, Canada
| | - Mark Henkelman
- Mouse Imaging Centre, Toronto Center of Phenogenomics, Toronto, Ontario, Canada
- Neurosciences and Mental Health Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Andreas Schulze
- Genetics and Genome Biology Program, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada
- Department of Pediatrics, University of Toronto, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
50
|
Klinke G, Richter S, Monostori P, Schmidt-Mader B, García-Cazorla A, Artuch R, Christ S, Opladen T, Hoffmann GF, Blau N, Okun JG. Targeted cerebrospinal fluid analysis for inborn errors of metabolism on an LC-MS/MS analysis platform. J Inherit Metab Dis 2020; 43:712-725. [PMID: 31930732 DOI: 10.1002/jimd.12213] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/13/2019] [Revised: 12/20/2019] [Accepted: 01/10/2020] [Indexed: 12/27/2022]
Abstract
BACKGROUND Laboratory investigations of cerebrospinal fluid (CSF) are essential when suspecting an inborn error of metabolism (IEM) involving neurological features. Available tests are currently performed on different analytical platforms, requiring a large sample volume and long turnaround time, which often delays timely diagnosis. Therefore, it would be preferable to have an "one-instrument" targeted multi-metabolite approach. METHOD A liquid chromatography-tandem mass spectrometry (LC-MS/MS) platform, based on two different methods for analysing 38 metabolites using positive and negative electrospray ionisation modes, was established. To allow for platform extension, both methods were designed to use the same CSF sample preparation procedure and to be run on the same separation column (ACE C18-PFP). RESULTS Assessment of the LC-MS/MS platform methods was first made by analytical validation, followed by the establishment of literature-based CSF cut-off values and reference ranges, and by the measurement of available samples obtained from patients with confirmed diagnoses of aromatic l-amino acid decarboxylase deficiency, guanidinoacetate methyltransferase deficiency, ornithine aminotransferase deficiency, cerebral folate deficiency and methylenetetrahydrofolate reductase deficiency. CONCLUSION An extendable targeted LC-MS/MS platform was developed for the analysis of multiple metabolites in CSF, thereby distinguishing samples from patients with IEM from non-IEM samples. Reference concentrations for several biomarkers in CSF are provided for the first time. By measurement on a single analytical platform, less sample volume is required (200 μL), diagnostic results are obtained faster, and preanalytical issues are reduced. SYNOPSIS LC-MS/MS platform for CSF analysis consisting of two differentially designed methods.
Collapse
Affiliation(s)
- Glynis Klinke
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Sylvia Richter
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Péter Monostori
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Brigitte Schmidt-Mader
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Angels García-Cazorla
- Department of Clinical Biochemistry and Pediatric Neurology, Institut de Recerca Sant Joan de Déu, Center for Biomedical Research on Rare Diseases (CIBERER), Barcelona, Spain
| | - Rafael Artuch
- Department of Clinical Biochemistry and Pediatric Neurology, Institut de Recerca Sant Joan de Déu, Center for Biomedical Research on Rare Diseases (CIBERER), Barcelona, Spain
| | - Stine Christ
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Thomas Opladen
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Georg F Hoffmann
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Nenad Blau
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| | - Jürgen G Okun
- Department of General Pediatrics, Division of Neuropediatrics and Metabolic Medicine, Centre for Pediatric and Adolescent Medicine, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|