Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 287
Filter
1.
Cell ; 175(1): 239-253.e17, 2018 09 20.
Article in English | MEDLINE | ID: mdl-30197081

ABSTRACT

Many disease-causing missense mutations affect intrinsically disordered regions (IDRs) of proteins, but the molecular mechanism of their pathogenicity is enigmatic. Here, we employ a peptide-based proteomic screen to investigate the impact of mutations in IDRs on protein-protein interactions. We find that mutations in disordered cytosolic regions of three transmembrane proteins (GLUT1, ITPR1, and CACNA1H) lead to an increased clathrin binding. All three mutations create dileucine motifs known to mediate clathrin-dependent trafficking. Follow-up experiments on GLUT1 (SLC2A1), the glucose transporter causative of GLUT1 deficiency syndrome, revealed that the mutated protein mislocalizes to intracellular compartments. Mutant GLUT1 interacts with adaptor proteins (APs) in vitro, and knocking down AP-2 reverts the cellular mislocalization and restores glucose transport. A systematic analysis of other known disease-causing variants revealed a significant and specific overrepresentation of gained dileucine motifs in structurally disordered cytosolic domains of transmembrane proteins. Thus, several mutations in disordered regions appear to cause "dileucineopathies."


Subject(s)
Glucose Transporter Type 1/physiology , Intrinsically Disordered Proteins/genetics , Intrinsically Disordered Proteins/physiology , Amino Acid Motifs/genetics , Amino Acid Sequence , Animals , Binding Sites , Calcium Channels, T-Type/genetics , Calcium Channels, T-Type/physiology , Carbohydrate Metabolism, Inborn Errors , Clathrin/metabolism , Cytoplasm/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Inositol 1,4,5-Trisphosphate Receptors/genetics , Inositol 1,4,5-Trisphosphate Receptors/physiology , Intrinsically Disordered Proteins/metabolism , Leucine/metabolism , Membrane Proteins/metabolism , Mice , Mice, Inbred C57BL , Monosaccharide Transport Proteins/deficiency , Mutation/genetics , Peptides , Protein Binding , Proteomics/methods
2.
Neurogenetics ; 25(2): 69-78, 2024 Apr.
Article in English | MEDLINE | ID: mdl-38190079

ABSTRACT

Glucose transporter type 1 deficiency syndrome (GLUT-1DS) is characterized by alterations in glucose translocation through the blood-brain barrier (BBB) due to mutation involving the GLUT-1 transporter. The fundamental therapy is ketogenic diet (KD) that provide an alternative energetic substrate - ketone bodies that across the BBB via MCT-1 - for the brain. Symptoms are various and include intractable seizure, acquired microcephalia, abnormal ocular movement, movement disorder, and neurodevelopment delay secondary to an energetic crisis for persistent neuroglycopenia. KD is extremely effective in controlling epileptic seizures and has a positive impact on movement disorders and cognitive impairment. Cases of KD resistance are rare, and only a few of them are reported in the literature, all regarding seizure. Our study describes a peculiar case of GLUT-1DS due to a new deletion involving the first codon of SLC2A1 gene determining a loss of function with a resistance to KD admitted to hospital due to intractable episodes of dystonia. This patient presented a worsening of symptomatology at higher ketonemia values but without hyperketosis and showed a complete resolution of symptomatology while maintaining low ketonemia values. Our study proposes an in-silico genomic and proteomic analysis aimed at explaining the atypical response to KD exhibited by our patient. In this way, we propose a new clinical and research approach based on precision medicine and molecular modelling to be applied to patients with GLUT-1DS resistant to first-line treatment with ketogenic diet by in silico study of genetic and altered protein product.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Diet, Ketogenic , Glucose Transporter Type 1 , Monosaccharide Transport Proteins/deficiency , Humans , Glucose Transporter Type 1/genetics , Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/diet therapy , Carbohydrate Metabolism, Inborn Errors/diagnosis , Male , Female , Computer Simulation
3.
Mov Disord ; 39(8): 1386-1396, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38725190

ABSTRACT

BACKGROUND: Paroxysmal movement disorders are common in Glut1 deficiency syndrome (Glut1DS). Not all patients respond to or tolerate ketogenic diets. OBJECTIVES: The objective was to evaluate the effectiveness and safety of triheptanoin in reducing the frequency of disabling movement disorders in patients with Glut1DS not receiving a ketogenic diet. METHODS: UX007G-CL301 was a randomized, double-blind, placebo-controlled, phase 3 crossover study. After a 6-week run-in, eligible patients were randomized 1:1 to the first sequence (triheptanoin/placebo or placebo/triheptanoin) titration plus maintenance, followed by washout and the opposite sequence titration plus maintenance. The placebo (safflower oil) matched the appearance, taste, and smell of triheptanoin. Open-label triheptanoin was administered in the extension. The frequency of disabling paroxysmal movement disorder events per 4 weeks (recorded by diary during maintenance; primary endpoint) was assessed by Wilcoxon rank-sum test. RESULTS: Forty-three patients (children, n = 16; adults, n = 27) were randomized and treated. There was no difference between triheptanoin and placebo in the mean (interquartile range) number of disabling paroxysmal movement disorder events (14.3 [4.7-38.3] vs. 11.8; [3.2-28.7]; Hodges-Lehmann estimated median difference: 1.46; 95% confidence interval, -1.12 to 4.36; P = 0.2684). Treatment-emergent adverse events were mild/moderate in severity and included diarrhea, vomiting, upper abdominal pain, headache, and nausea. Two patients discontinued the study because of non-serious adverse events that were predominantly gastrointestinal. The study was closed early during the open-label extension because of lack of effectiveness. Seven patients continued to receive triheptanoin compassionately. CONCLUSION: There were no significant differences between the triheptanoin and placebo groups in the frequency of disabling movement disorder events during the double-blind maintenance period. © 2024 The Authors. Movement Disorders published by Wiley Periodicals LLC on behalf of International Parkinson and Movement Disorder Society.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Cross-Over Studies , Humans , Female , Male , Double-Blind Method , Carbohydrate Metabolism, Inborn Errors/drug therapy , Adolescent , Child , Child, Preschool , Adult , Young Adult , Monosaccharide Transport Proteins/deficiency , Movement Disorders/drug therapy , Treatment Outcome , Triglycerides
4.
Epilepsia ; 65(8): 2213-2226, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38767952

ABSTRACT

In glucose transporter 1 deficiency syndrome (Glut1DS), glucose transport into brain is reduced due to impaired Glut1 function in endothelial cells at the blood-brain barrier. This can lead to shortages of glucose in brain and is thought to contribute to seizures. Ketogenic diets are the first-line treatment and, among many beneficial effects, provide auxiliary fuel in the form of ketone bodies that are largely metabolized by neurons. However, Glut1 is also the main glucose transporter in astrocytes. Here, we review data indicating that glucose shortage may also impact astrocytes in addition to neurons and discuss the expected negative biochemical consequences of compromised astrocytic glucose transport for neurons. Based on these effects, auxiliary fuels are needed for both cell types and adding medium chain triglycerides (MCTs) to ketogenic diets is a biochemically superior treatment for Glut1DS compared to classical ketogenic diets. MCTs provide medium chain fatty acids (MCFAs), which are largely metabolized by astrocytes and not neurons. MCFAs supply energy and contribute carbons for glutamine and γ-aminobutyric acid synthesis, and decanoic acid can also block α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid glutamate receptors. MCTs do not compete with metabolism of ketone bodies mostly occurring in neurons. Triheptanoin, an anaplerotic but also gluconeogenic uneven MCT, may be another potential addition to ketogenic diets, although maintenance of "ketosis" can be difficult. Gene therapy has also targeted both endothelial cells and astrocytes. Other approaches to increase fuel delivery to the brain currently investigated include exchange of Glut1DS erythrocytes with healthy cells, infusion of lactate, and pharmacological improvement of glucose transport. In conclusion, although it remains difficult to assess impaired astrocytic energy metabolism in vivo, astrocytic energy needs are most likely not met by ketogenic diets in Glut1DS. Thus, we propose prospective studies including monitoring of blood MCFA levels to find optimal doses for add-on MCT to ketogenic diets and assessing of short- and long-term outcomes.


Subject(s)
Astrocytes , Carbohydrate Metabolism, Inborn Errors , Diet, Ketogenic , Energy Metabolism , Glucose Transporter Type 1 , Astrocytes/metabolism , Humans , Carbohydrate Metabolism, Inborn Errors/metabolism , Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/diet therapy , Energy Metabolism/physiology , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Glucose/metabolism , Animals , Monosaccharide Transport Proteins/deficiency , Monosaccharide Transport Proteins/metabolism , Monosaccharide Transport Proteins/genetics
5.
Eur J Neurol ; 31(8): e16325, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38803061

ABSTRACT

BACKGROUND AND PURPOSE: Glucose transporter-1 (GLUT1) deficiency syndrome (GLUT1-DS) is a metabolic disorder due to reduced expression of GLUT1, a glucose transporter of the central nervous system. GLUT1-DS is caused by heterozygous SLC2A1 variants that mostly arise de novo. Here, we report a large family with heterogeneous phenotypes related to a novel SLC2A1 variant. METHODS: We present clinical and genetic features of a five-generation family with GLUT1-DS. RESULTS: The 14 (nine living) affected members had heterogeneous phenotypes, including seizures (11/14), behavioral disturbances (5/14), mild intellectual disability (3/14), and/or gait disabilities (2/14). Brain magnetic resonance imaging revealed hippocampal sclerosis in the 8-year-old proband, who also had drug-responsive absences associated with attention-deficit/hyperactivity disorder. His 52-year-old father, who had focal epilepsy since childhood, developed paraparesis related to a reversible myelitis associated with hypoglycorrhachia. Molecular study detected a novel heterozygous missense variant (c.446C>T) in exon 4 of SLC2A1 (NM: 006516.2) that cosegregated with the illness. This variant causes an amino acid replacement (p.Pro149Leu) at the fourth transmembrane segment of GLUT1, an important domain located at its catalytic core. CONCLUSIONS: Our study illustrates the extremely heterogenous phenotypes in familial GLUT1-DS, ranging from milder classic phenotypes to more subtle neurological disorder including paraparesis. This novel SLC2A1 variant (c.446C>T) provides new insight into the pathophysiology of GLUT1-DS.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Glucose Transporter Type 1 , Pedigree , Phenotype , Child , Female , Humans , Male , Middle Aged , Carbohydrate Metabolism, Inborn Errors/genetics , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/deficiency , Magnetic Resonance Imaging , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/deficiency , Mutation, Missense/genetics
6.
Dev Med Child Neurol ; 66(11): 1466-1475, 2024 Nov.
Article in English | MEDLINE | ID: mdl-38655597

ABSTRACT

AIM: To investigate the clinical characteristics of non-epileptic seizures due to transient brain dysfunction caused by energy deficiency after prolonged fasting or exercise in individuals with glucose transporter type 1 deficiency syndrome (Glut1DS), and then elucidate further the seizure features to distinguish non-epileptic seizures from epileptic seizures. METHOD: This retrospective case-control study included 57 non-epileptic seizures and 23 epileptic seizures (control group) in 14 individuals (11 males, three females; aged 5-44 years, median = 20 years) with Glut1DS, all with a heterozygous pathogenic SLC2A1 mutation. RESULTS: Non-epileptic seizures were classified as paroxysmal altered consciousness (n = 8), movement disorders (n = 35) (eye-head movements, ataxia, spasticity, weakness, involuntary movement), dysaesthesia (n = 8), and vomiting (n = 6) at the peak ages at onset of 5 to 10 years. Ketogenic diet therapy was effective in 33 of 43 (77%) non-epileptic seizures. Providing supplementary food before high-impact exercise or during attacks prevented or mitigated non-epileptic seizures in some individuals. Glut1DS-associated non-epileptic seizures are fundamentally situation-related seizures with specific provoking and ameliorating factors. Non-epileptic seizures can be distinguished from epileptic seizures by the absence of complete consciousness loss and rapid postictal recovery despite prolonged seizures. INTERPRETATION: Non-epileptic seizures are not well recognized but require different therapeutic approaches compared to epileptic seizures. Awareness of the differentiation of non-epileptic seizures from epileptic seizures is essential when performing preventive or therapeutic decision-making for acute exacerbation seizures. WHAT THIS PAPER ADDS: Non-epileptic seizures are invariably situation-related seizures. Non-epileptic seizures were classified as altered consciousness, movement disorders, dysaesthesia, and vomiting. Non-epileptic seizures were characterized by the absence of complete consciousness loss and were accompanied by rapid recovery. Non-epileptic seizures can occur simultaneously or consecutively with another. Supplementary food can be effective in preventing the development of sustained exercise-induced movement disorders.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Epilepsy , Monosaccharide Transport Proteins , Seizures , Humans , Female , Male , Child , Adolescent , Carbohydrate Metabolism, Inborn Errors/complications , Carbohydrate Metabolism, Inborn Errors/diagnosis , Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/physiopathology , Seizures/diagnosis , Seizures/etiology , Child, Preschool , Retrospective Studies , Monosaccharide Transport Proteins/deficiency , Monosaccharide Transport Proteins/genetics , Case-Control Studies , Young Adult , Adult , Epilepsy/diagnosis , Epilepsy/complications , Diet, Ketogenic , Diagnosis, Differential , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/deficiency
7.
Eur J Pediatr ; 183(9): 3665-3678, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38954008

ABSTRACT

The purpose of this study is to investigate the diagnostic and prognostic role of cerebrospinal fluid (CSF) biomarkers in the diagnostic work-up of glucose transporter 1 (GLUT1) deficiency. Reported here is a systematic review according to PRISMA guidelines collecting clinical and biochemical data about all published patients who underwent CSF analysis. Clinical phenotypes were compared between groups defined by the levels of CSF glucose (≤ 2.2 mmol/L versus > 2.2 mmol/L), CSF/blood glucose ratio (≤ 0.45 versus > 0.45), and CSF lactate (≤ 1 mmol/L versus > 1 mmol/L). Five hundred sixty-two patients fulfilled the inclusion criteria with a mean age at the diagnosis of 8.6 ± 6.7 years. Patients with CSF glucose ≤ 2.2 mmol/L and CSF/blood glucose ratio ≤ 0.45 presented with an earlier onset of symptoms (16.4 ± 22.0 versus 54.4 ± 45.9 months, p < 0.01; 15.7 ± 23.8 versus 40.9 ± 38.0 months, p < 0.01) and received an earlier molecular genetic confirmation (92.1 ± 72.8 versus 157.1 ± 106.2 months, p < 0.01). CSF glucose ≤ 2.2 mmol/L was consistently associated with response to ketogenic diet (p = 0.018) and antiseizure medications (p = 0.025). CSF/blood glucose ratio ≤ 0.45 was significantly associated with absence seizures (p = 0.048), paroxysmal exercise-induced dyskinesia (p = 0.046), and intellectual disability (p = 0.016) while CSF lactate > 1 mmol/L was associated with a response to antiseizure medications (p = 0.026) but not to ketogenic diet.Conclusions:This systematic review supported the diagnostic usefulness of lumbar puncture for the early identification of patients with GLUT1 deficiency responsive to treatments especially if they present with co-occurring epilepsy, movement, and neurodevelopmental disorders. What is Known: • Phenotypes of GLUT1 deficiency syndrome range between early epileptic and developmental encephalopathy to paroxysmal movement disorders and developmental impairment What is New: • CSF blood/glucose ratio may predict better than CSF glucose the diagnosis in children presenting with early onset absences • CSF blood/glucose ratio may predict better than CSF glucose the diagnosis in children presenting with paroxysmal exercise induced dyskinesia and intellectual disability. • CSF glucose may predict better than CSF blood/glucose and lactate the response to ketogenic diet and antiseizure medications.


Subject(s)
Biomarkers , Carbohydrate Metabolism, Inborn Errors , Humans , Biomarkers/cerebrospinal fluid , Biomarkers/blood , Prognosis , Carbohydrate Metabolism, Inborn Errors/diagnosis , Carbohydrate Metabolism, Inborn Errors/cerebrospinal fluid , Child , Lactic Acid/cerebrospinal fluid , Lactic Acid/blood , Blood Glucose/analysis , Diet, Ketogenic , Child, Preschool , Glucose Transporter Type 1/genetics , Glucose/cerebrospinal fluid , Monosaccharide Transport Proteins/deficiency
8.
Neurol Sci ; 45(9): 4539-4547, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38589768

ABSTRACT

OBJECTIVE: The aim of the study is to analyze microstate patterns in GLUT1-DS, both before and after the ketogenic diet (KD). METHODS: We conducted microstate analysis of a patient with GLUT-1 DS and 27 healthy controls. A systematic literature review and meta-analysis was done. We compared the parameters of the patients with those of healthy controls and the incorporating findings in literature. RESULTS: The durations of the patient were notably shorter, and the occurrence rates were longer than those of healthy controls and incorporating findings from the review. After 10 months of KD, the patient's microstate durations exhibited an increase from 53.05 ms, 57.17 ms, 61.80 ms, and 49.49 ms to 60.53 ms, 63.27 ms, 71.11 ms, and 66.55 ms. The occurrence rates changed from 4.0774 Hz, 4.9462 Hz, 4.8006 Hz, and 4.0579 Hz to 3.3354 Hz, 3.7893 Hz, 3.5956 Hz, and 4.1672 Hz. In healthy controls, the durations of microstate class A, B, C, and D were 61.86 ms, 63.58 ms, 70.57 ms, and 72.00 ms, respectively. CONCLUSIONS: Our findings suggest EEG microstates may be a promising biomarker for monitoring the effect of KD. Administration of KD may normalize the dysfunctional patterns of temporal parameters.


Subject(s)
Diet, Ketogenic , Electroencephalography , Humans , Carbohydrate Metabolism, Inborn Errors/diet therapy , Carbohydrate Metabolism, Inborn Errors/diagnosis , Carbohydrate Metabolism, Inborn Errors/physiopathology , Biomarkers/blood , Female , Monosaccharide Transport Proteins/deficiency , Male , Prospective Studies
9.
J Hum Nutr Diet ; 37(4): 827-846, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38838079

ABSTRACT

BACKGROUND: The ketogenic diet (KD) is a high fat, moderate protein and very low carbohydrate diet. It can be used as a medical treatment for drug-resistant epilepsy (DRE), glucose transporter 1 deficiency syndrome and pyruvate dehydrogenase deficiency. The aim of this scoping review was to map the KD literature, with a focus on epilepsy and associated metabolic conditions, to summarise the current evidence-base and identify any gaps. METHODS: This review was conducted using JBI scoping review methodological guidance and the PRISMA extension for scoping reviews reporting guidance. A comprehensive literature search was conducted in September 2021 and updated in February 2024 using MEDLINE, CINAHL, AMED, EmBASE, CAB Abstracts, Scopus and Food Science Source databases. RESULTS: The initial search yielded 2721 studies and ultimately, data were extracted from 320 studies that fulfilled inclusion criteria for the review. There were five qualitative studies, and the remainder were quantitative, including 23 randomised controlled trials (RCTs) and seven quasi-experimental studies. The USA published the highest number of KD studies followed by China, South Korea and the UK. Most studies focused on the classical KD and DRE. The studies key findings suggest that the KD is efficacious, safe and tolerable. CONCLUSIONS: There are opportunities available to expand the scope of future KD research, particularly to conduct high-quality RCTs and further qualitative research focused on the child's needs and family support to improve the effectiveness of KDs.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Diet, Ketogenic , Drug Resistant Epilepsy , Pyruvate Dehydrogenase Complex Deficiency Disease , Humans , Diet, Ketogenic/methods , Pyruvate Dehydrogenase Complex Deficiency Disease/diet therapy , Child , Drug Resistant Epilepsy/diet therapy , Carbohydrate Metabolism, Inborn Errors/diet therapy , Monosaccharide Transport Proteins/deficiency , Child, Preschool , Male , Female , Adolescent
10.
Mol Cell ; 58(5): 711-2, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-26046643

ABSTRACT

In this issue, Lee et al. (2015) show that PKC directly phosphorylates the glucose transporter Glut1, in order to promote glucose uptake in response to growth factor signaling.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/genetics , Glucose Transporter Type 1/metabolism , Monosaccharide Transport Proteins/deficiency , Protein Kinase C-alpha/physiology , Animals , Humans
11.
Mol Cell ; 58(5): 845-53, 2015 Jun 04.
Article in English | MEDLINE | ID: mdl-25982116

ABSTRACT

Protein kinase C has been implicated in the phosphorylation of the erythrocyte/brain glucose transporter, GLUT1, without a clear understanding of the site(s) of phosphorylation and the possible effects on glucose transport. Through in vitro kinase assays, mass spectrometry, and phosphospecific antibodies, we identify serine 226 in GLUT1 as a PKC phosphorylation site. Phosphorylation of S226 is required for the rapid increase in glucose uptake and enhanced cell surface localization of GLUT1 induced by the phorbol ester 12-O-tetradecanoyl-phorbol-13-acetate (TPA). Endogenous GLUT1 is phosphorylated on S226 in primary endothelial cells in response to TPA or VEGF. Several naturally occurring, pathogenic mutations that cause GLUT1 deficiency syndrome disrupt this PKC phosphomotif, impair the phosphorylation of S226 in vitro, and block TPA-mediated increases in glucose uptake. We demonstrate that the phosphorylation of GLUT1 on S226 regulates glucose transport and propose that this modification is important in the physiological regulation of glucose transport.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/genetics , Glucose Transporter Type 1/metabolism , Monosaccharide Transport Proteins/deficiency , Protein Kinase C-alpha/physiology , Amino Acid Sequence , Animals , Biological Transport , Carbohydrate Metabolism, Inborn Errors/enzymology , Cell Line , Endothelial Cells/metabolism , Erythrocytes/metabolism , Glucose/metabolism , Glucose Transporter Type 1/genetics , HeLa Cells , Humans , Molecular Sequence Data , Monosaccharide Transport Proteins/genetics , Mutation, Missense , Phosphorylation , Protein Processing, Post-Translational , Rats , Xenopus laevis
12.
J Neurochem ; 162(6): 483-500, 2022 09.
Article in English | MEDLINE | ID: mdl-35943296

ABSTRACT

Glucose is an important source of energy for the central nervous system. Its uptake at the blood-brain barrier (BBB) is mostly mediated via glucose transporter 1 (GLUT1), a facilitated transporter encoded by the SLC2A1 gene. GLUT1 Deficiency Syndrome (GLUT1DS) is a haploinsufficiency characterized by mutations in the SLC2A1 gene, resulting in impaired glucose uptake at the BBB and clinically characterized by epileptic seizures and movement disorder. A major limitation is an absence of in vitro models of the BBB reproducing the disease. This study aimed to characterize an in vitro model of GLUT1DS using human pluripotent stem cells (iPSCs). Two GLUT1DS clones were generated (GLUT1-iPSC) from their original parental clone iPS(IMR90)-c4 by CRISPR/Cas9 and differentiated into brain microvascular endothelial cells (iBMECs). Cells were characterized in terms of SLC2A1 expression, changes in the barrier function, glucose uptake and metabolism, and angiogenesis. GLUT1DS iPSCs and iBMECs showed comparable phenotype to their parental control, with exception of reduced GLUT1 expression at the protein level. Although no major disruption in the barrier function was reported in the two clones, a significant reduction in glucose uptake accompanied by an increase in glycolysis and mitochondrial respiration was reported in both GLUT1DS-iBMECs. Finally, impaired angiogenic features were reported in such clones compared to the parental clone. Our study provides the first documented characterization of GLUT1DS-iBMECs generated by CRISPR-Cas9, suggesting that GLUT1 truncation appears detrimental to brain angiogenesis and brain endothelial bioenergetics, but maybe not be detrimental to iBMECs differentiation and barriergenesis. Our future direction is to further characterize the functional outcome of such truncated product, as well as its impact on other cells of the neurovascular unit.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Induced Pluripotent Stem Cells , Monosaccharide Transport Proteins , Blood-Brain Barrier/metabolism , Endothelial Cells/metabolism , Glucose/metabolism , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Humans , Induced Pluripotent Stem Cells/metabolism , Monosaccharide Transport Proteins/deficiency
13.
Clin Genet ; 102(1): 40-55, 2022 07.
Article in English | MEDLINE | ID: mdl-35388452

ABSTRACT

Glucose transporter 1 deficiency syndrome (GLUT1DS) is a neurometabolic disorder caused by haploinsufficiency of the GLUT1 glucose transporter (encoded by SLC2A1) leading to defective glucose transport across the blood-brain barrier. This work describes the genetic analysis of 56 patients with clinical or biochemical GLUT1DS hallmarks. 55.4% of these patients had a pathogenic variant of SLC2A1, and 23.2% had a variant in one of 13 different genes. No pathogenic variant was identified for the remaining patients. Expression analysis of SLC2A1 indicated a reduction in SLC2A1 mRNA in patients with pathogenic variants of this gene, as well as in one patient with a pathogenic variant in SLC9A6, and in three for whom no candidate variant was identified. Thus, the clinical and biochemical hallmarks generally associated with GLUT1DS may be caused by defects in genes other than SLC2A1.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Carbohydrate Metabolism, Inborn Errors/genetics , Genetic Testing , Glucose Transporter Type 1/genetics , Humans , Monosaccharide Transport Proteins/deficiency , Monosaccharide Transport Proteins/genetics
14.
Epilepsia ; 63(7): 1748-1760, 2022 07.
Article in English | MEDLINE | ID: mdl-35441706

ABSTRACT

OBJECTIVE: This study was undertaken to evaluate efficacy and long-term safety of triheptanoin in patients >1 year old, not on a ketogenic diet, with drug-resistant seizures associated with glucose transporter 1 deficiency syndrome (Glut1DS). METHODS: UX007G-CL201 was a randomized, double-blind, placebo-controlled trial. Following a 6-week baseline period, eligible patients were randomized 3:1 to triheptanoin or placebo. Dosing was titrated to 35% of total daily calories over 2 weeks. After an 8-week placebo-controlled period, all patients received open-label triheptanoin through Week 52. RESULTS: The study included 36 patients (15 children, 13 adolescents, eight adults). A median 12.6% reduction in overall seizure frequency was observed in the triheptanoin arm relative to baseline, and a 13.5% difference was observed relative to placebo (p = .58). In patients with absence seizures only (n = 9), a median 62.2% reduction in seizure frequency was observed in the triheptanoin arm relative to baseline. Only one patient with absence seizures only was present in the control group, preventing comparison. No statistically significant differences in seizure frequency were observed. Common treatment-emergent adverse events included diarrhea, vomiting, abdominal pain, and nausea, mostly mild or moderate in severity. No serious adverse events were considered to be treatment related. One patient discontinued due to status epilepticus. SIGNIFICANCE: Triheptanoin did not significantly reduce seizure frequency in patients with Glut1DS not on the ketogenic diet. Treatment was associated with mild to moderate gastrointestinal treatment-related events; most resolved following dose reduction or interruption and/or medication for treatment. Triheptanoin was not associated with any long-term safety concerns when administered at dose levels up to 35% of total daily caloric intake for up to 1 year.


Subject(s)
Drug Resistant Epilepsy , Epilepsy, Absence , Triglycerides , Adolescent , Adult , Anticonvulsants/therapeutic use , Carbohydrate Metabolism, Inborn Errors , Child , Double-Blind Method , Drug Resistant Epilepsy/drug therapy , Drug Therapy, Combination , Epilepsy, Absence/drug therapy , Glucose Transporter Type 1/genetics , Humans , Monosaccharide Transport Proteins/deficiency , Seizures/drug therapy , Treatment Outcome , Triglycerides/therapeutic use
15.
Neuropediatrics ; 53(2): 129-132, 2022 04.
Article in English | MEDLINE | ID: mdl-34674205

ABSTRACT

Glucose transporter type-1 deficiency syndrome (Glut1 DS) is a rare disorder with various manifestations. Early diagnosis is crucial because treatment with the ketogenic diet can lead to clinical improvement. Here, we report the cases of two siblings with Glut1 DS and one of them presented with sleep disorder which is a rare and atypical manifestation of Glut1 DS. Patient 1 was a 3.5-year-old boy who presented with paroxysmal loss of tone and weakness of the whole body with unresponsiveness after waking up. He also had excessive daytime sleepiness, insomnia, and restless sleep. His other clinical findings included focal seizures, paroxysmal exercise-induced dyskinesia (PED), ataxia, mild global developmental delay, and hyperactivity. Patient 2 was a 5.5-year-old boy who presented with drug-resistant focal epilepsy, global developmental delay, paroxysmal dystonia, and ataxia. A novel heterozygous nonsense variant of SLC2A1, c.1177G > T (p.Glu393*), classified as a pathogenic variant, was identified in both patients, but not in their parents' blood. After treatment with the modified Atkins diet, their neurological functions significantly improved. In conclusion, we reported two siblings with variable phenotypes of Glut1 DS with a novel nonsense mutation. Although sleep disorder and daytime somnolence were the nonclassical manifestations of Glut1 DS, the diagnostic evaluation of possible Glut1 DS in patients presented with daytime sleepiness, particularly in cases with the cooccurrence of seizures or movement disorders should be considered.


Subject(s)
Carbohydrate Metabolism, Inborn Errors , Chorea , Diet, Ketogenic , Epilepsy , Sleep Wake Disorders , Ataxia/etiology , Ataxia/genetics , Carbohydrate Metabolism, Inborn Errors/complications , Carbohydrate Metabolism, Inborn Errors/diagnosis , Carbohydrate Metabolism, Inborn Errors/genetics , Child, Preschool , Chorea/genetics , Epilepsy/genetics , Glucose Transporter Type 1/genetics , Humans , Male , Monosaccharide Transport Proteins/deficiency , Mutation , Seizures , Sleep Wake Disorders/genetics
16.
Zhonghua Yi Xue Yi Chuan Xue Za Zhi ; 39(8): 884-888, 2022 Aug 10.
Article in Zh | MEDLINE | ID: mdl-35929942

ABSTRACT

OBJECTIVE: To analyze the clinical phenotype and variant of SLC2A1 gene in a Chinese pedigree affected with glucose transporter type 1 deficiency syndrome (GLUT1-DS). METHODS: Clinical data of a child who was treated due to delayed motor and language development and his family members were collected. DNA was extracted from peripheral blood samples and subjected to high-throughput medical exome sequencing. Candidate variant was verified by Sanger sequencing of his parents and sister. The genotype-phenotype correlation was explored. RESULTS: The child, his mother and sister had common manifestations such as delayed mental and motor development, poor exercise tolerance, easy fatigue and paroxysmal dystonia, but the difference was that the child and his mother had microcephaly and seizures, while his sister did not. A heterozygous missense SLC2A1 c.191T>C (p.L64P) variant was identified in all affected members, which was unreported previously. CONCLUSION: The missense SLC2A1 c.191T>C (p.L64P) variant probably underlay the disease in the proband and his mother and sister. Variability of the clinical phenotypes has reflected the genetic and phenotypic diversity of GLUT1-DS. Detection of the novel variant has enriched the spectrum of GLUT1-DS mutations.


Subject(s)
Pedigree , Carbohydrate Metabolism, Inborn Errors , China , Glucose Transporter Type 1/genetics , Humans , Monosaccharide Transport Proteins/deficiency , Mutation , Phenotype
17.
Pflugers Arch ; 472(9): 1299-1343, 2020 09.
Article in English | MEDLINE | ID: mdl-32789766

ABSTRACT

Energy demand of neurons in brain that is covered by glucose supply from the blood is ensured by glucose transporters in capillaries and brain cells. In brain, the facilitative diffusion glucose transporters GLUT1-6 and GLUT8, and the Na+-D-glucose cotransporters SGLT1 are expressed. The glucose transporters mediate uptake of D-glucose across the blood-brain barrier and delivery of D-glucose to astrocytes and neurons. They are critically involved in regulatory adaptations to varying energy demands in response to differing neuronal activities and glucose supply. In this review, a comprehensive overview about verified and proposed roles of cerebral glucose transporters during health and diseases is presented. Our current knowledge is mainly based on experiments performed in rodents. First, the functional properties of human glucose transporters expressed in brain and their cerebral locations are described. Thereafter, proposed physiological functions of GLUT1, GLUT2, GLUT3, GLUT4, and SGLT1 for energy supply to neurons, glucose sensing, central regulation of glucohomeostasis, and feeding behavior are compiled, and their roles in learning and memory formation are discussed. In addition, diseases are described in which functional changes of cerebral glucose transporters are relevant. These are GLUT1 deficiency syndrome (GLUT1-SD), diabetes mellitus, Alzheimer's disease (AD), stroke, and traumatic brain injury (TBI). GLUT1-SD is caused by defect mutations in GLUT1. Diabetes and AD are associated with changed expression of glucose transporters in brain, and transporter-related energy deficiency of neurons may contribute to pathogenesis of AD. Stroke and TBI are associated with changes of glucose transporter expression that influence clinical outcome.


Subject(s)
Alzheimer Disease/metabolism , Brain/metabolism , Carbohydrate Metabolism, Inborn Errors/metabolism , Diabetes Mellitus/metabolism , Glucose Transport Proteins, Facilitative/metabolism , Monosaccharide Transport Proteins/deficiency , Animals , Glucose Transport Proteins, Facilitative/genetics , Humans , Monosaccharide Transport Proteins/metabolism
18.
Pflugers Arch ; 472(9): 1371-1383, 2020 09.
Article in English | MEDLINE | ID: mdl-32474749

ABSTRACT

Glucose transport is intimately linked to red blood cell physiology. Glucose is the unique energy source for these cells, and defects in glucose metabolism or transport activity are associated with impaired red blood cell morphology and deformability leading to reduced lifespan. In vertebrate erythrocytes, glucose transport is mediated by GLUT1 (in humans) or GLUT4 transporters. These proteins also account for dehydroascorbic acid (DHA) transport through erythrocyte membrane. The peculiarities of glucose transporters and the red blood cell pathologies involving GLUT1 are summarized in the present review.


Subject(s)
Anemia, Hemolytic/metabolism , Carbohydrate Metabolism, Inborn Errors/metabolism , Erythrocytes/metabolism , Glucose Transporter Type 1/metabolism , Monosaccharide Transport Proteins/deficiency , Anemia, Hemolytic/genetics , Animals , Carbohydrate Metabolism, Inborn Errors/genetics , Glucose Transporter Type 1/genetics , Humans , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism
19.
J Membr Biol ; 253(2): 87-99, 2020 04.
Article in English | MEDLINE | ID: mdl-32025761

ABSTRACT

Human sodium-independent glucose cotransporter 1 (hGLUT1) has been studied for its tetramerization and multimerization at the cell surface. Homozygous or compound heterozygous mutations in hGLUT1 elicit GLUT1-deficiency syndrome (GLUT1-DS), a metabolic disorder, which results in impaired glucose transport into the brain. The reduced cell surface expression or loss of function have been shown for some GLUT1 mutants. However, the mechanism by which deleterious mutations affect protein structure, conformational stability and GLUT1 oligomerization is not known and require investigation. In this review, we combined previous knowledge of GLUT1 mutations with hGLUT1 crystal structure to analyze native interactions and several natural single-point mutations. The modeling of native hGLUT1 structure confirmed the roles of native residues in forming a range of side-chain interactions. Interestingly, the modeled mutants pointed to the formation of a variety of non-native novel interactions, altering interaction networks and potentially eliciting protein misfolding. Self-aggregation of the last part of hGLUT1 was predicted using protein aggregation prediction tool. Furthermore, an increase in aggregation potential in the aggregation-prone regions was estimated for several mutants suggesting increased aggregation of misfolded protein. Protein stability change analysis predicted that GLUT1 mutant proteins are unstable. Combining GLUT1 oligomerization behavior with our modeling, aggregation prediction, and protein stability analyses, this work provides state-of-the-art view of GLUT1 genetic mutations that could destabilize native interactions, generate novel interactions, trigger protein misfolding, and enhance protein aggregation in a disease state.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/genetics , Carbohydrate Metabolism, Inborn Errors/metabolism , Genetic Variation , Glucose Transporter Type 1/genetics , Glucose Transporter Type 1/metabolism , Monosaccharide Transport Proteins/deficiency , Amino Acid Substitution , Animals , Carbohydrate Metabolism, Inborn Errors/diagnosis , Glucose Transporter Type 1/chemistry , Humans , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Monosaccharide Transport Proteins/genetics , Monosaccharide Transport Proteins/metabolism , Mutation , Polymorphism, Single Nucleotide , Protein Aggregates , Protein Binding , Protein Conformation , Protein Stability , Structure-Activity Relationship
20.
J Inherit Metab Dis ; 43(2): 216-222, 2020 03.
Article in English | MEDLINE | ID: mdl-31605543

ABSTRACT

Glucose transporter type 1 deficiency syndrome (GLUT1DS) is characterised by deficient glucose transport over the blood-brain barrier and reduced glucose availability in the brain. This causes epilepsy, movement disorders, and cognitive impairment. Treatment with ketogenic diet provides ketones as alternative energy source. However, not all GLUT1DS patients are on dietary treatment (worldwide registry: 77/181 [43%] of patients). The current 25-year experience allows evaluation of effects and tolerability of dietary treatment for GLUT1DS. To this end, literature was searched up to January 2019 for individual case reports and series reporting (side) effects of dietary treatment for GLUT1DS. Upon aggregation of data for analysis, we identified 270 GLUT1DS patients with dietary treatment with a mean follow-up of 53 months. Epilepsy improved for 83% of 230 patients and remained unchanged for 17%, movement disorders improved for 82% of 127 patients and remained unchanged for 17%, and cognition improved for 59% of 58 patients and remained stable for 40%. Effects on epilepsy were seen within days/weeks and were most pronounced in patients with early treatment initiation. Effects on movement disorders were noticed within months and were strongest in patients with higher cerebrospinal fluid-to-blood glucose ratio. Although side effects were minimal, 18% of 270 patients reported poor compliance. In individual patients, symptoms deteriorated upon low ketosis, poor compliance, or treatment discontinuation. Based on the good tolerability and strong favourable effect of dietary treatment on GLUT1DS symptoms, we advocate dietary treatment in all GLUT1DS patients and prompt diagnosis or screening to allow early treatment.


Subject(s)
Carbohydrate Metabolism, Inborn Errors/diet therapy , Cognitive Dysfunction/diet therapy , Diet, Ketogenic , Epilepsy/diet therapy , Monosaccharide Transport Proteins/deficiency , Movement Disorders/diet therapy , Cognitive Dysfunction/etiology , Epilepsy/etiology , Humans , Movement Disorders/etiology
SELECTION OF CITATIONS
SEARCH DETAIL