Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 197
1.
Cell Mol Life Sci ; 81(1): 186, 2024 Apr 17.
Article En | MEDLINE | ID: mdl-38632116

Pathogenic variants in SLC6A8, the gene which encodes creatine transporter SLC6A8, prevent creatine uptake in the brain and result in a variable degree of intellectual disability, behavioral disorders (e.g., autism spectrum disorder), epilepsy, and severe speech and language delay. There are no treatments to improve neurodevelopmental outcomes for creatine transporter deficiency (CTD). In this spotlight, we summarize recent advances in innovative molecules to treat CTD, with a focus on dodecyl creatine ester, the most promising drug candidate.


Autism Spectrum Disorder , Brain Diseases, Metabolic, Inborn , Creatine/deficiency , Intellectual Disability , Mental Retardation, X-Linked , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Humans , Creatine/genetics , Creatine/therapeutic use , Brain Diseases, Metabolic, Inborn/drug therapy , Brain Diseases, Metabolic, Inborn/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/drug therapy , Mental Retardation, X-Linked/genetics
2.
Neurology ; 102(8): e209243, 2024 Apr 23.
Article En | MEDLINE | ID: mdl-38531017

BACKGROUND AND OBJECTIVES: Creatine transporter deficiency (CTD) is a rare X-linked genetic disorder characterized by intellectual disability (ID). We evaluated the clinical characteristics and trajectory of patients with CTD and the impact of the disease on caregivers to identify relevant endpoints for future therapeutic trials. METHODS: As part of a French National Research Program, patients with CTD were included based on (1) a pathogenic SLC6A8 variant and (2) ID and/or autism spectrum disorder. Families and patients were referred by the physician who ordered the genetic analysis through Reference Centers of ID from rare causes and inherited metabolic diseases. After we informed the patients and their parents/guardians about the study, all of them gave written consent and were included. A control group of age-matched and sex-matched patients with Fragile X syndrome was also included. Physical examination, neuropsychological assessments, and caregiver impact were assessed. All data were analyzed using R software. RESULTS: Thirty-one patients (27 male, 4 female) were included (25/31 aged 18 years or younger). Most of the patients (71%) had symptoms at <24 months of age. The mean age at diagnosis was 6.5 years. Epilepsy occurred in 45% (mean age at onset: 8 years). Early-onset behavioral disorder occurred in 82%. Developmental trajectory was consistently delayed (fine and gross motor skills, language, and communication/sociability). Half of the patients with CTD had axial hypotonia during the first year of life. All patients were able to walk without help, but 7/31 had ataxia and only 14/31 could walk tandem gait. Most of them had abnormal fine motor skills (27/31), and most of them had language impairment (30/31), but 12/23 male patients (52.2%) completed the Peabody Picture Vocabulary Test. Approximately half (14/31) had slender build. Most of them needed nursing care (20/31), generally 1-4 h/d. Adaptive assessment (Vineland) confirmed that male patients with CTD had moderate-to-severe ID. Most caregivers (79%) were at risk of burnout, as shown by Caregiver Burden Inventory (CBI) > 36 (significantly higher than for patients with Fragile X syndrome) with a high burden of time dependence. DISCUSSION: In addition to clinical endpoints, such as the assessment of epilepsy and the developmental trajectory of the patient, the Vineland scale, PPVT5, and CBI are of particular interest as outcome measures for future trials. TRIAL REGISTRATION INFORMATION: ANSM Registration Number 2010-A00327-32.


Autism Spectrum Disorder , Brain Diseases, Metabolic, Inborn , Creatine/deficiency , Epilepsy , Fragile X Syndrome , Intellectual Disability , Mental Retardation, X-Linked , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Humans , Male , Female , Child , Caregiver Burden , Nerve Tissue Proteins
3.
Mol Genet Metab ; 142(1): 108362, 2024 May.
Article En | MEDLINE | ID: mdl-38452609

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.


Amidinotransferases , Amidinotransferases/deficiency , Amino Acid Metabolism, Inborn Errors , Creatine , Creatine/deficiency , Guanidinoacetate N-Methyltransferase , Intellectual Disability , Language Development Disorders , Movement Disorders/congenital , Nerve Tissue Proteins , Plasma Membrane Neurotransmitter Transport Proteins , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Speech Disorders , Humans , Guanidinoacetate N-Methyltransferase/deficiency , Guanidinoacetate N-Methyltransferase/genetics , Creatine/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Amidinotransferases/genetics , Amidinotransferases/metabolism , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/diagnosis , Mutation , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/diagnosis , Phenotype , Data Curation , Developmental Disabilities
4.
Mol Genet Metab ; 142(1): 108455, 2024 May.
Article En | MEDLINE | ID: mdl-38531184

Creatine transporter deficiency has been described with normal or uninformative levels of creatine and creatinine in plasma, while urine has been the preferred specimen type for biochemical diagnosis. We report a cohort of untreated patients with creatine transporter deficiency and abnormal plasma creatine panel results, characterized mainly by markedly decreased plasma creatinine. We conclude that plasma should be considered a viable specimen type for the biochemical diagnosis of this disorder, and abnormal results should be followed up with further confirmatory testing.


Brain Diseases, Metabolic, Inborn , Creatine , Creatine/deficiency , Creatinine , Mental Retardation, X-Linked , Plasma Membrane Neurotransmitter Transport Proteins , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Humans , Creatine/blood , Creatine/urine , Creatinine/blood , Creatinine/urine , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/blood , Male , Female , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/blood , Mental Retardation, X-Linked/diagnosis , Child , Child, Preschool , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/blood , Nerve Tissue Proteins/deficiency , Infant , Adolescent , Membrane Transport Proteins/genetics , Membrane Transport Proteins/deficiency , Membrane Transport Proteins/blood , Adult
5.
J Mol Biol ; 436(2): 168383, 2024 01 15.
Article En | MEDLINE | ID: mdl-38070861

Creatine is an essential metabolite for the storage and rapid supply of energy in muscle and nerve cells. In humans, impaired metabolism, transport, and distribution of creatine throughout tissues can cause varying forms of mental disability, also known as creatine deficiency syndrome (CDS). So far, 80 mutations in the creatine transporter (SLC6A8) have been associated to CDS. To better understand the effect of human genetic variants on the physiology of SLC6A8 and their possible impact on CDS, we studied 30 missense variants including 15 variants of unknown significance, two of which are reported here for the first time. We expressed these variants in HEK293 cells and explored their subcellular localization and transport activity. We also applied computational methods to predict variant effect and estimate site-specific changes in thermodynamic stability. To explore variants that might have a differential effect on the transporter's conformers along the transport cycle, we constructed homology models of the inward facing, and outward facing conformations. In addition, we used mass-spectrometry to study proteins that interact with wild type SLC6A8 and five selected variants in HEK293 cells. In silico models of the protein complexes revealed how two variants impact the interaction interface of SLC6A8 with other proteins and how pathogenic variants lead to an enrichment of ER protein partners. Overall, our integrated analysis disambiguates the pathogenicity of 15 variants of unknown significance revealing diverse mechanisms of pathogenicity, including two previously unreported variants obtained from patients suffering from the creatine deficiency syndrome.


Brain Diseases, Metabolic, Inborn , Creatine , Mental Retardation, X-Linked , Nerve Tissue Proteins , Plasma Membrane Neurotransmitter Transport Proteins , Humans , Creatine/deficiency , HEK293 Cells , Mental Retardation, X-Linked/genetics , Nerve Tissue Proteins/deficiency , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Brain Diseases, Metabolic, Inborn/genetics , DNA Mutational Analysis/methods , Mutation, Missense , Computational Biology/methods
6.
Am J Med Genet A ; 194(2): 337-345, 2024 Feb.
Article En | MEDLINE | ID: mdl-37850681

Seizures occur in up to 59% of boys with creatine transporter deficiency (CTD). While seizure phenotypes have been previously described, electroencephalogram (EEG) findings have only been reported in several case reports. In this prospective observational study, we report seizure characteristics and EEG findings in combination with neurobehavioral and SLC6A8 pathogenic variants in twenty males with CTD. Eighteen study participants (SP) underwent video-EEG, and seven had follow-up EEG recordings. Seizures typically occurred by age of 2 years. Thirteen (65%) had non-febrile seizures, requiring anti-seizure medications in nine. Four had febrile seizures. Seizures were bilateral tonic-clonic in 7 SP and focal impaired awareness in 5 SP; often responding to 1 to 2 antiseizure medications. EEG showed slowing in 5 SP, beta activity in 6 SP, and focal/multifocal, and/or generalized epileptiform activity in 9 SP. Follow-up EEGs in 7 SP showed emergence of epileptiform activity in 1 SP, and increased activity in 2 SP. In conclusion, seizures were frequent in our cohort but tended to respond to antiseizure medications. Longitudinal follow up provided further insight into emergence of seizures and EEG abnormalities soliciting future studies with long term follow up. Biomarkers of epileptogenicity in CTD are needed to predict seizures in this population.


Brain Diseases, Metabolic, Inborn , Creatine/deficiency , Electroencephalography , Mental Retardation, X-Linked , Male , Humans , Child, Preschool , Mutation , Seizures/diagnosis , Seizures/drug therapy , Seizures/genetics , Nerve Tissue Proteins , Plasma Membrane Neurotransmitter Transport Proteins/genetics
7.
Clin Chim Acta ; 532: 29-36, 2022 Jul 01.
Article En | MEDLINE | ID: mdl-35588794

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.


Brain Diseases, Metabolic, Inborn , Guanidinoacetate N-Methyltransferase , Intellectual Disability , Nerve Tissue Proteins , Plasma Membrane Neurotransmitter Transport Proteins , Brain Diseases, Metabolic, Inborn/diagnosis , Brain Diseases, Metabolic, Inborn/genetics , Child , Creatine/deficiency , Guanidinoacetate N-Methyltransferase/genetics , Humans , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Syndrome
8.
Brain Dev ; 44(4): 271-280, 2022 Apr.
Article En | MEDLINE | ID: mdl-34974949

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.


Brain Diseases, Metabolic, Inborn/diagnosis , Creatine/deficiency , Guanidinoacetate N-Methyltransferase/deficiency , Language Development Disorders/diagnosis , Mental Retardation, X-Linked/diagnosis , Movement Disorders/congenital , Neurodevelopmental Disorders/diagnosis , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Brain Diseases, Metabolic, Inborn/complications , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/physiopathology , Child , Child, Preschool , Creatine/genetics , Female , Follow-Up Studies , Guanidinoacetate N-Methyltransferase/genetics , Humans , India , Language Development Disorders/complications , Language Development Disorders/genetics , Language Development Disorders/physiopathology , Male , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/physiopathology , Movement Disorders/complications , Movement Disorders/diagnosis , Movement Disorders/genetics , Movement Disorders/physiopathology , Neurodevelopmental Disorders/etiology , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/physiopathology , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Retrospective Studies
9.
Genes (Basel) ; 12(8)2021 07 24.
Article En | MEDLINE | ID: mdl-34440297

Creatine (Cr) Transporter Deficiency (CTD) is an X-linked metabolic disorder, mostly caused by missense mutations in the SLC6A8 gene and presenting with intellectual disability, autistic behavior, and epilepsy. There is no effective treatment for CTD and patients need lifelong assistance. Thus, the research of novel intervention strategies is a major scientific challenge. Animal models are an excellent tool to dissect the disease pathogenetic mechanisms and drive the preclinical development of therapeutics. This review illustrates the current knowledge about Cr metabolism and CTD clinical aspects, with a focus on mainstay diagnostic and therapeutic options. Then, we discuss the rodent models of CTD characterized in the last decade, comparing the phenotypes expressed within clinically relevant domains and the timeline of symptom development. This analysis highlights that animals with the ubiquitous deletion/mutation of SLC6A8 genes well recapitulate the early onset and the complex pathological phenotype of the human condition. Thus, they should represent the preferred model for preclinical efficacy studies. On the other hand, brain- and cell-specific conditional mutants are ideal for understanding the basis of CTD at a cellular and molecular level. Finally, we explain how CTD models might provide novel insight about the pathogenesis of other disorders, including cancer.


Brain Diseases, Metabolic, Inborn/pathology , Brain Diseases, Metabolic, Inborn/therapy , Central Nervous System/pathology , Creatine/deficiency , Disease Models, Animal , Mental Retardation, X-Linked/pathology , Mental Retardation, X-Linked/therapy , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Animals , Biomarkers/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Creatine/metabolism , Humans , Mental Retardation, X-Linked/metabolism , Mice , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Rats
10.
JCI Insight ; 6(17)2021 09 08.
Article En | MEDLINE | ID: mdl-34324436

The creatine transporter (CrT) maintains brain creatine (Cr) levels, but the effects of its deficiency on energetics adaptation under stress remain unclear. There are also no effective treatments for CrT deficiency, the second most common cause of X-linked intellectual disabilities. Herein, we examined the consequences of CrT deficiency in brain energetics and stress-adaptation responses plus the effects of intranasal Cr supplementation. We found that CrT-deficient (CrT-/y) mice harbored dendritic spine and synaptic dysgenesis. Nurtured newborn CrT-/y mice maintained baseline brain ATP levels, with a trend toward signaling imbalance between the p-AMPK/autophagy and mTOR pathways. Starvation elevated the signaling imbalance and reduced brain ATP levels in P3 CrT-/y mice. Similarly, CrT-/y neurons and P10 CrT-/y mice showed an imbalance between autophagy and mTOR signaling pathways and greater susceptibility to cerebral hypoxia-ischemia and ischemic insults. Notably, intranasal administration of Cr after cerebral ischemia increased the brain Cr/N-acetylaspartate ratio, partially averted the signaling imbalance, and reduced infarct size more potently than intraperitoneal Cr injection. These findings suggest important functions for CrT and Cr in preserving the homeostasis of brain energetics in stress conditions. Moreover, intranasal Cr supplementation may be an effective treatment for congenital CrT deficiency and acute brain injury.


Brain Diseases, Metabolic, Inborn/genetics , Brain/metabolism , Creatine/deficiency , DNA/genetics , Membrane Transport Proteins/genetics , Mental Retardation, X-Linked/genetics , Mutation , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Animals , Animals, Newborn , Brain/ultrastructure , Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic, Inborn/pathology , Creatine/genetics , Creatine/metabolism , DNA Mutational Analysis , Disease Models, Animal , Homeostasis , Male , Membrane Transport Proteins/deficiency , Mental Retardation, X-Linked/metabolism , Mental Retardation, X-Linked/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Mutant Strains , Microscopy, Electron , Neurons/metabolism , Neurons/ultrastructure , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/metabolism
11.
Regul Toxicol Pharmacol ; 123: 104939, 2021 Jul.
Article En | MEDLINE | ID: mdl-33961952

In standard general toxicology studies in two species to support clinical development, cyclocreatine, a creatine analog for the treatment of creatine transporter deficiency, caused deaths, convulsions, and/or multi-organ pathology. The potential translatability of these findings to patients was evaluated by comparing toxicity of cyclocreatine in wild-type mice to creatine transporter-deficient mice, a model of the human disease. A biodistribution study indicated greater accumulation of cyclocreatine in the brains of wild-type mice, consistent with its ability to be transported by the creatine transporter. Subsequent toxicology studies confirmed greater sensitivity of wild-type mice to cyclocreatine-induced toxicity. Exposure at the no observed adverse effect level in creatine transporter-deficient (554 µg*hr/ml) mice exceeded exposure at the maximum tolerated dose in wild-type (248 µg*hr/ml) mice. When dosed at 300 mg/kg/day for 3 months, cyclocreatine-related mortality, convulsions, and multi-organ pathology were observed in wild-type mice whereas there were no adverse findings in creatine transporter-deficient mice. Brain vacuolation was common to both strains. Although transporter-deficient mice appeared to be more sensitive, the finding had no functional correlates in this strain. The results highlight the importance of considering models of disease for toxicology in cases where they may be relevant to assessing safety in the intended patient population.


Antineoplastic Agents/toxicity , Creatinine/analogs & derivatives , Disease Models, Animal , Animals , Brain , Brain Diseases, Metabolic, Inborn , Creatine/deficiency , Creatinine/toxicity , Humans , Membrane Transport Proteins , Mental Retardation, X-Linked , Mice , No-Observed-Adverse-Effect Level , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Seizures , Tissue Distribution
12.
Genet Med ; 23(10): 1864-1872, 2021 10.
Article En | MEDLINE | ID: mdl-34050321

PURPOSE: Creatine transporter deficiency (CTD) is a rare X-linked disorder of creatine transport caused by pathogenic variants in SLC6A8 (Xq28). CTD features include developmental delay, seizures, and autism spectrum disorder. This study was designed to investigate CTD cardiac phenotype and sudden death risk. METHODS: We performed a cross-sectional analysis of CTD males between 2017 and 2020. Subjects underwent evaluation with electrocardiogram (ECG), echocardiography, and ambulatory ECG with comparable analysis in creatine transporter deficient mice (Slc6a8-/y) using ECG, echocardiography, exercise testing, and indirect calorimetry. RESULTS: Eighteen subjects with CTD (18 males, age 7.4 [3.8] years) were evaluated: seven subjects (39%) had QTc ≥ 470 milliseconds: 510.3 ± 29.0 vs. 448.3 ± 15.9, P < 0.0001. The QTc ≥ 470 milliseconds cohort had increased left ventricular internal dimension (diastole) ([LVIDd] Z-score: 0.22 ± 0.74, n = 7 vs. -0.93 ± 1.0, n = 11, P = 0.0059), and diminished left ventricular posterior wall dimension (diastole) ([LVPWDd, in mm]: 5.0 ± 0.6, n = 7 vs. 5.7 ± 0.8, n = 11, P = 0.0183), when compared to subjects with normal or borderline QTc prolongation. Similar ECG and echocardiographic abnormalities were seen in Slc6a8-/y mice. Additionally, Slc6a8-/y mice had diminished survival (65%). CONCLUSION: Prolonged QTc and abnormal echocardiographic parameters consistent with developing cardiomyopathy are seen in some male subjects with CTD. Slc6a8-/y mice recapitulated these cardiac abnormalities. Male CTD subjects may be at increased risk for cardiac dysfunction and sudden death.


Autism Spectrum Disorder , Creatine , Animals , Brain Diseases, Metabolic, Inborn , Creatine/deficiency , Cross-Sectional Studies , Death, Sudden , Humans , Male , Mental Retardation, X-Linked , Mice , Plasma Membrane Neurotransmitter Transport Proteins/deficiency
13.
Sci Rep ; 11(1): 1636, 2021 01 15.
Article En | MEDLINE | ID: mdl-33452333

Creatine is an organic compound used as fast phosphate energy buffer to recycle ATP, important in tissues with high energy demand such as muscle or brain. Creatine is taken from the diet or endogenously synthetized by the enzymes AGAT and GAMT, and specifically taken up by the transporter SLC6A8. Deficit in the endogenous synthesis or in the transport leads to Cerebral Creatine Deficiency Syndromes (CCDS). CCDS are characterized by brain creatine deficiency, intellectual disability with severe speech delay, behavioral troubles such as attention deficits and/or autistic features, and epilepsy. Among CCDS, the X-linked creatine transporter deficiency (CTD) is the most prevalent with no efficient treatment so far. Different mouse models of CTD were generated by doing long deletions in the Slc6a8 gene showing reduced brain creatine and cognitive deficiencies or impaired motor function. We present a new knock-in (KI) rat model of CTD holding an identical point mutation found in patients with reported lack of transporter activity. KI males showed brain creatine deficiency, increased urinary creatine/creatinine ratio, cognitive deficits and autistic-like traits. The Slc6a8Y389C KI rat fairly enriches the spectrum of CTD models and provides new data about the pathology, being the first animal model of CTD carrying a point mutation.


Brain/metabolism , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Animals , Base Sequence , Behavior, Animal , Body Weight , Brain Diseases, Metabolic, Inborn/genetics , Brain Diseases, Metabolic, Inborn/pathology , Creatine/blood , Creatine/deficiency , Creatine/genetics , Disease Models, Animal , Female , Gene Knock-In Techniques , Genotype , Humans , Male , Memory, Short-Term , Mental Retardation, X-Linked/genetics , Mental Retardation, X-Linked/pathology , Mutation, Missense , Nerve Tissue Proteins/chemistry , Nerve Tissue Proteins/metabolism , Plasma Membrane Neurotransmitter Transport Proteins/chemistry , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Plasma Membrane Neurotransmitter Transport Proteins/metabolism , Rats
14.
J Inherit Metab Dis ; 44(4): 939-948, 2021 07.
Article En | MEDLINE | ID: mdl-33389772

The severe impact on brain function and lack of effective therapy for patients with creatine (Cr) transporter deficiency motivated the generation of three ubiquitous Slc6a8 deficient mice (-/y). While each mouse knock-out line has similar behavioral effects at 2 to 3 months of age, other features critical to the efficient use of these mice in drug discovery are unclear or lacking: the concentration of Cr in brain and heart differ widely between mouse lines, there are limited data on histopathologic changes, and no data on Cr uptake. Here, we determined survival, measured endogenous Cr and uptake of its deuterium-labeled analogue Cr-d3 using a liquid chromatography coupled with tandem mass spectrometry assay, and performed comprehensive histopathologic examination on the Slc6a8-/y mouse developed by Skelton et al. Our results show that Slc6a8-/y mice have widely varying organ-specific uptake of Cr-d3, significantly diminished growth with the exception of brain, progressive vacuolar myopathy, and markedly shortened lifespan.


Brain Diseases, Metabolic, Inborn/genetics , Creatine/deficiency , Mental Retardation, X-Linked/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Animals , Brain Diseases, Metabolic, Inborn/pathology , Chromatography, Liquid , Creatine/genetics , Mental Retardation, X-Linked/pathology , Mice , Mice, Knockout , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Tandem Mass Spectrometry
16.
Am J Physiol Heart Circ Physiol ; 320(2): H613-H629, 2021 02 01.
Article En | MEDLINE | ID: mdl-33337958

Creatine kinase (CK) is considered the main phosphotransfer system in the heart, important for overcoming diffusion restrictions and regulating mitochondrial respiration. It is substrate limited in creatine-deficient mice lacking l-arginine:glycine amidinotransferase (AGAT) or guanidinoacetate N-methyltranferase (GAMT). Our aim was to determine the expression, activity, and mitochondrial coupling of hexokinase (HK) and adenylate kinase (AK), as these represent alternative energy transfer systems. In permeabilized cardiomyocytes, we assessed how much endogenous ADP generated by HK, AK, or CK stimulated mitochondrial respiration and how much was channeled to mitochondria. In whole heart homogenates, and cytosolic and mitochondrial fractions, we measured the activities of AK, CK, and HK. Lastly, we assessed the expression of the major HK, AK, and CK isoforms. Overall, respiration stimulated by HK, AK, and CK was ∼25, 90, and 80%, respectively, of the maximal respiration rate, and ∼20, 0, and 25%, respectively, was channeled to the mitochondria. The activity, distribution, and expression of HK, AK, and CK did not change in GAMT knockout (KO) mice. In AGAT KO mice, we found no changes in AK, but we found a higher HK activity in the mitochondrial fraction, greater expression of HK I, but a lower stimulation of respiration by HK. Our findings suggest that mouse hearts depend less on phosphotransfer systems to facilitate ADP flux across the mitochondrial membrane. In AGAT KO mice, which are a model of pure creatine deficiency, the changes in HK may reflect changes in metabolism as well as influence mitochondrial regulation and reactive oxygen species production.NEW & NOTEWORTHY In creatine-deficient AGAT-/- and GAMT-/- mice, the myocardial creatine kinase system is substrate limited. It is unknown whether subcellular localization and mitochondrial ADP channeling by hexokinase and adenylate kinase may compensate as alternative phosphotransfer systems. Our results show no changes in adenylate kinase, which is the main alternative to creatine kinase in heart. However, we found increased expression and activity of hexokinase I in AGAT-/- cardiomyocytes. This could affect mitochondrial regulation and reactive oxygen species production.


Amidinotransferases/deficiency , Amino Acid Metabolism, Inborn Errors/enzymology , Creatine/deficiency , Energy Metabolism , Guanidinoacetate N-Methyltransferase/deficiency , Hexokinase/metabolism , Intellectual Disability/enzymology , Language Development Disorders/enzymology , Mitochondria, Heart/enzymology , Movement Disorders/congenital , Myocytes, Cardiac/enzymology , Speech Disorders/enzymology , Adenosine Diphosphate/metabolism , Adenylate Kinase/metabolism , Amidinotransferases/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Animals , Cell Respiration , Creatine Kinase/metabolism , Developmental Disabilities/enzymology , Developmental Disabilities/genetics , Disease Models, Animal , Female , Guanidinoacetate N-Methyltransferase/genetics , Intellectual Disability/genetics , Language Development Disorders/genetics , Male , Mice, Inbred C57BL , Mice, Knockout , Movement Disorders/enzymology , Movement Disorders/genetics , Speech Disorders/genetics
17.
Gene ; 768: 145260, 2021 Feb 05.
Article En | MEDLINE | ID: mdl-33164824

Correct diagnosis of children presenting with developmental delay and intellectual disability remains challenging due to the complex and heterogeneous etiology. High throughput sequencing technologies like exome sequencing have become more commonly available and are significantly improving genetic testing. We present two siblings - a 14-year old male and an 8-year old female patient - with a similar clinical phenotype that was characterized by combined developmental delay primarily affecting speech, mild to moderate intellectual disability, behavioral abnormalities, and autism spectrum disorder, but with no congenital anomalies. The sister showed additional muscular hypotonia and more pronounced dysmorphic features compared to her brother. Both parents had psychiatric disorders and mild to moderate intellectual disability. A common genetic etiology in the siblings was suspected. Metabolic, psychological and neuroradiological examinations were complemented by basic genetic testing including chromosome analysis and array comparative genomics hybridization analysis (CGH), followed by exome sequencing and combined data analysis of the family. Exome sequencing identified two different underlying genetic conditions: in the sister, a maternally inherited pathogenic variant c.1661C > T, p.Pro554Leu in SLC6A8 (NM_005629.4) was identified causing cerebral creatine deficiency syndrome 1 (MIM #300352) which was confirmed by MR spectroscopy and treated accordingly. In the brother, a paternally inherited 16p13.11 duplication was identified by exome sequencing and considered to be likely associated with his and possibly his father's phenotype. The 16p13.11 duplication had been previously identified in an array CGH but had not been prioritized due to the lack of segregation in the siblings. In conclusion, we report a case of intra-familial locus heterogeneity of developmental delay in two siblings. We advocate for the need of unbiased and comprehensive genetic testing to provide accurate diagnosis despite locus heterogeneity.


Autism Spectrum Disorder/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/genetics , Brain Diseases, Metabolic, Inborn/genetics , Child , Chromosomes, Human, Pair 16/genetics , Comparative Genomic Hybridization , Creatine/deficiency , Creatine/genetics , Female , Gene Duplication/genetics , Genetic Testing , Humans , Male , Mental Retardation, X-Linked/genetics , Muscle Hypotonia/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Siblings , Exome Sequencing
18.
BMJ Case Rep ; 13(12)2020 Dec 17.
Article En | MEDLINE | ID: mdl-33334757

X-linked creatine transporter deficiency is caused by the deficiency of the creatine transporter encoded by the SLC6A8 gene on Xq28. We here report a 3-year-old boy with global developmental delay, autism and epilepsy. He had a normal MRI of the brain. Brain magnetic resonance spectroscopy (MRS) subsequently showed an abnormally small creatine peak. His high urine creatine/creatinine ratio further suggested the diagnosis, later confirmed by hemizygous mutation detected in the SLC6A8 gene. His mother was also heterozygous for the same mutation. Supplementation with creatine monohydrate, arginine, and glycine (precursors of creatine) and supportive therapies, resulted in modest clinical improvement after 12 months. This case highlights the importance of doing MRS for boys with global delay/intellectual disability, autism and epilepsy even with a normal MRI of the brain, to pick up a potentially treatable cause.


Autistic Disorder/genetics , Brain Diseases, Metabolic, Inborn/diagnosis , Creatine/deficiency , Epilepsy/genetics , Intellectual Disability/genetics , Mental Retardation, X-Linked/diagnosis , Nerve Tissue Proteins/genetics , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Brain Diseases, Metabolic, Inborn/complications , Brain Diseases, Metabolic, Inborn/genetics , Child, Preschool , Creatine/analysis , Creatine/genetics , Creatine/metabolism , Hemizygote , Humans , Magnetic Resonance Spectroscopy , Male , Mental Retardation, X-Linked/complications , Mental Retardation, X-Linked/genetics , Mutation , Nerve Tissue Proteins/deficiency , Plasma Membrane Neurotransmitter Transport Proteins/genetics
19.
Am J Intellect Dev Disabil ; 125(6): 475-480, 2020 11 01.
Article En | MEDLINE | ID: mdl-33211814

Although norm-referenced scores are essential to the identification of disability, they possess several features which affect their sensitivity to change. Norm-referenced scores often decrease over time among people with neurodevelopmental disorders who exhibit slower-than-average increases in ability. Further, the reliability of norm-referenced scores is lower at the tails of the distribution, resulting in floor effects and increased measurement error for people with neurodevelopmental disorders. In contrast, the person ability scores generated during the process of constructing a standardized test with item response theory are designed to assess change. We illustrate these limitations of norm-referenced scores, and relative advantages of ability scores, using data from studies of autism spectrum disorder and creatine transporter deficiency.


Neurodevelopmental Disorders/diagnosis , Neuropsychological Tests/standards , Outcome Assessment, Health Care/standards , Psychometrics/standards , Autism Spectrum Disorder/diagnosis , Brain Diseases, Metabolic, Inborn/diagnosis , Child , Creatine/deficiency , Humans , Mental Retardation, X-Linked/diagnosis , Plasma Membrane Neurotransmitter Transport Proteins/deficiency
20.
Sci Rep ; 10(1): 18361, 2020 10 27.
Article En | MEDLINE | ID: mdl-33110151

Creatine Transporter Deficiency (CTD) is an inborn error of metabolism presenting with intellectual disability, behavioral disturbances and epilepsy. There is currently no cure for this disorder. Here, we employed novel biomarkers for monitoring brain function, together with well-established behavioral readouts for CTD mice, to longitudinally study the therapeutic efficacy of cyclocreatine (cCr) at the preclinical level. Our results show that cCr treatment is able to partially correct hemodynamic responses and EEG abnormalities, improve cognitive deficits, revert autistic-like behaviors and protect against seizures. This study provides encouraging data to support the potential therapeutic benefit of cyclocreatine or other chemically modified lipophilic analogs of Cr.


Autistic Disorder/etiology , Brain Diseases, Metabolic, Inborn/drug therapy , Cognition Disorders/etiology , Creatine/deficiency , Creatinine/analogs & derivatives , Epilepsy/etiology , Mental Retardation, X-Linked/drug therapy , Plasma Membrane Neurotransmitter Transport Proteins/deficiency , Animals , Autistic Disorder/drug therapy , Blood-Brain Barrier , Brain Diseases, Metabolic, Inborn/complications , Cerebrovascular Circulation/drug effects , Cognition Disorders/drug therapy , Creatinine/therapeutic use , Disease Models, Animal , Electroencephalography , Epilepsy/drug therapy , Hemodynamics/drug effects , Male , Mental Retardation, X-Linked/complications , Mice , Mice, Inbred C57BL , Phenotype , Seizures/drug therapy , Seizures/etiology , Stereotyped Behavior/drug effects
...