Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 29
1.
PLoS Genet ; 17(2): e1009307, 2021 02.
Article En | MEDLINE | ID: mdl-33524012

Hundreds of mutations in a single gene result in rare diseases, but why mutations induce severe or attenuated states remains poorly understood. Defect in glycine decarboxylase (GLDC) causes Non-ketotic Hyperglycinemia (NKH), a neurological disease associated with elevation of plasma glycine. We unified a human multiparametric NKH mutation scale that separates severe from attenuated neurological disease with new in silico tools for murine and human genome level-analyses, gathered in vivo evidence from mice engineered with top-ranking attenuated and a highly pathogenic mutation, and integrated the data in a model of pre- and post-natal disease outcomes, relevant for over a hundred major and minor neurogenic mutations. Our findings suggest that highly severe neurogenic mutations predict fatal, prenatal disease that can be remedied by metabolic supplementation of dams, without amelioration of persistent plasma glycine. The work also provides a systems approach to identify functional consequences of mutations across hundreds of genetic diseases. Our studies provide a new framework for a large scale understanding of mutation functions and the prediction that severity of a neurogenic mutation is a direct measure of pre-natal disease in neurometabolic NKH mouse models. This framework can be extended to analyses of hundreds of monogenetic rare disorders where the underlying genes are known but understanding of the vast majority of mutations and why and how they cause disease, has yet to be realized.


Disease Models, Animal , Glycine Dehydrogenase (Decarboxylating)/chemistry , Glycine Dehydrogenase (Decarboxylating)/genetics , Glycine/metabolism , Hyperglycinemia, Nonketotic/genetics , Animals , Female , Genomics , Genotype , Glycine/genetics , Humans , Hyperglycinemia, Nonketotic/metabolism , Hyperglycinemia, Nonketotic/pathology , Male , Mass Spectrometry , Mice , Mice, Inbred C57BL , Mice, Knockout , Mutation , Mutation, Missense , Phenotype
2.
Sci Rep ; 11(1): 3148, 2021 02 04.
Article En | MEDLINE | ID: mdl-33542258

Delayed emergence from anesthesia was previously reported in a case study of a child with Glycine Encephalopathy. To investigate the neural basis of this delayed emergence, we developed a zebrafish glial glycine transporter (glyt1 - / -) mutant model. We compared locomotor behaviors; dose-response curves for tricaine, ketamine, and 2,6-diisopropylphenol (propofol); time to emergence from these anesthetics; and time to emergence from propofol after craniotomy in glyt1-/- mutants and their siblings. To identify differentially active brain regions in glyt1-/- mutants, we used pERK immunohistochemistry as a proxy for brain-wide neuronal activity. We show that glyt1-/- mutants initiated normal bouts of movement less frequently indicating lethargy-like behaviors. Despite similar anesthesia dose-response curves, glyt1-/- mutants took over twice as long as their siblings to emerge from ketamine or propofol, mimicking findings from the human case study. Reducing glycine levels rescued timely emergence in glyt1-/- mutants, pointing to a causal role for elevated glycine. Brain-wide pERK staining showed elevated activity in hypnotic brain regions in glyt1-/- mutants under baseline conditions and a delay in sensorimotor integration during emergence from anesthesia. Our study links elevated activity in preoptic brain regions and reduced sensorimotor integration to lethargy-like behaviors and delayed emergence from propofol in glyt1-/- mutants.


Delayed Emergence from Anesthesia/genetics , Glycine Plasma Membrane Transport Proteins/genetics , Glycine/metabolism , Hyperglycinemia, Nonketotic/genetics , Neurons/metabolism , Preoptic Area/metabolism , Zebrafish Proteins/genetics , Aminobenzoates , Anesthesia, General , Anesthetics , Animals , Animals, Genetically Modified , Craniotomy , Delayed Emergence from Anesthesia/metabolism , Delayed Emergence from Anesthesia/physiopathology , Delayed Emergence from Anesthesia/prevention & control , Disease Models, Animal , Gene Expression , Glycine/pharmacology , Glycine Plasma Membrane Transport Proteins/deficiency , Hyperglycinemia, Nonketotic/drug therapy , Hyperglycinemia, Nonketotic/metabolism , Hyperglycinemia, Nonketotic/physiopathology , Ketamine , Locomotion/physiology , Neurons/drug effects , Neurons/pathology , Preoptic Area/drug effects , Preoptic Area/pathology , Propofol , Zebrafish , Zebrafish Proteins/deficiency , eIF-2 Kinase/genetics , eIF-2 Kinase/metabolism
3.
Rev. neurol. (Ed. impr.) ; 67(12): 491-504, 16 dic., 2018. ilus
Article Es | IBECS | ID: ibc-175179

Introducción. Los aminoácidos glutamato y glicina, aparte de su papel en la síntesis de proteínas, son dos neurotransmisores fundamentales en el sistema nervioso central de los mamíferos. El primero es ubicuo y está implicado en vías excitatorias de la neocorteza, la retina y el cerebelo, y el segundo está asociado a vías inhibitorias de zonas caudales del cerebro. Sin embargo, ambos comparten su manera de actuar al integrarse en el funcionamiento de los receptores de glutamato del tipo NMDA, fundamentales en la regulación de sistemas motores, sensitivos y cognitivos. Objetivo. Evidenciar la necesidad de una regulación exquisita de las concentraciones de glutamato y de glicina en los espacios intra y extracelulares del sistema nervioso mediante la actuación de transportadores muy específicos para ambos neurotransmisores localizados en la membrana plasmática de las neuronas y de las células de la glía. Desarrollo. Se describe el papel de los transportadores de glutamato y glicina en la neurotransmisión glutamatérgica y glicinérgica, y en el funcionamiento del sistema nervioso. Se señalan las consecuencias patológicas de los desequilibrios en estas vías de señalización. También se describe su participación en patologías como la esquizofrenia, el dolor crónico, la isquemia cerebral, la hiperplexia hereditaria, la hiperglicinemia no cetósica o trastornos neurodegenerativos. Conclusiones. El conocimiento de la forma molecular de actuar de los transportadores de glutamato y de glicina está permitiendo la identificación y el desarrollo de nuevas estrategias terapéuticas para patologías como las descritas y el desarrollo de nuevos fármacos


Introduction. The amino acids glutamate and glycine, apart from their role in protein synthesis, are two fundamental neurotransmitters in the central nervous system of mammals. The first one is ubiquitous and is involved in excitatory pathways of the neocortex, the retina and the cerebellum, and the second is involved in inhibitory pathways of brain caudal areas. However, both share their way of acting by integrating into the functioning of glutamate receptors of the NMDA type fundamentals in the regulation of motor, sensory and cognitive systems. Aim. To highlight the need for a fine regulation of glutamate and glycine concentrations in the intracellular and extracellular spaces of the nervous system through the action of very specific transporters for both neurotransmitters located in the plasma membrane of neurons and glial cells. Development. The role of the glutamate and glycine transporters in glutamatergic and glycinergic neurotransmission and in the functioning of the nervous system is described. The pathological consequences of imbalances in these signaling pathways are pointed out. We also describe its involvement in pathologies such as schizophrenia, chronic pain, cerebral ischemia, diseases such as hereditary hyperekplexia and the non-ketotic hyperglycinemia, and neurodegenerative disorders. Conclusions. The knowledge at molecular level of the way of acting of these transporters for glutamate and glycine is allowing the identification and development of new therapeutic strategies for pathologies such as those described above and the development of new drugs


Humans , Glutamic Acid , Glycine/metabolism , Central Nervous System/metabolism , Carrier Proteins/metabolism , Schizophrenia/metabolism , Chronic Pain/metabolism , Brain Ischemia/metabolism , Hyperglycinemia, Nonketotic/metabolism , Neurodegenerative Diseases/metabolism , Schizophrenia/physiopathology , Chronic Pain/physiopathology , Brain Ischemia/physiopathology , Hyperglycinemia, Nonketotic , Neurodegenerative Diseases/physiopathology
4.
J Theor Biol ; 455: 97-100, 2018 10 14.
Article En | MEDLINE | ID: mdl-30009793

Nonketotic-hyperglycinemia (NKH) is an autosomal recessive disorder associated with grave brain malformations and severe neurological symptoms, and also characterized by accumulation of a large amount of glycine in body fluids. NKH is caused by an inherited deficiency of the glycine cleavage system (GCS), which is the main system to degrade glycine in mammalians. These severe symptoms and grave bran malformations are not normally observed in the other amino acid metabolic disorders, suggesting that GCS should have unknown pivotal roles in brain development and function. Interestingly, GCS is indispensable in supplying proliferating cells with 5,10-methylenetetrahydrofolate as a one-carbon donor, which is essential for the synthesis of DNA in cell proliferation. Since GCS is expressed intensely and ubiquitously in the neuroepithelium, the lack of GCS might greatly impair the proliferation of neural stem cells. On the other hand, this system is also very important to regulate extracellular glycine concentrations. Since glycine is an important neurotransmitter, which binds to both glycine receptors and NMDA receptors, high glycine concentrations caused by the deficiency of GCS might cause the aberrant neurotransmission in the patient brains. Considering these unique two faces of GCS functions, proliferation disturbance and aberrant neurotransmission are intricately mixed in the developing brain, leading to the grave brain malformations and sever neurological symptoms.


Brain/metabolism , Glycine/metabolism , Hyperglycinemia, Nonketotic/metabolism , Models, Neurological , Nervous System Diseases/metabolism , Neural Stem Cells/metabolism , Brain/abnormalities , Brain/pathology , Cell Proliferation , DNA/genetics , Glycine/genetics , Humans , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/pathology , Nervous System Diseases/genetics , Nervous System Diseases/pathology , Neural Stem Cells/pathology
5.
Brain Dev ; 39(7): 601-605, 2017 Aug.
Article En | MEDLINE | ID: mdl-28325525

INTRODUCTION: Glycine encephalopathy (GE), also known as non-ketotic hyperglycinemia (NKH), is a rare inborn error of glycine metabolism caused by a defect in glycine cleavage system, a multi-enzyme complex located in mitochondrial membrane. This defect results in elevated glycine concentration in plasma and cerebrospinal fluid (CSF). Clinical manifestations vary from severe lethargy, hypoactivity and apneic episodes in the neonatal form, mild or moderate psychomotor delay and seizures in the infantile form, and abnormal behaviors, ataxia and choreoathetoid movements in late onset form. More than 50 GLDC mutations were found, reflecting large heterogeneity of the gene. METHODS: We describe the clinical, biochemical and molecular characteristics of three Palestinian siblings who have distinct clinical phenotypes. Molecular study was performed utilizing standard Polymerase Chain Reaction (PCR) amplification then direct DNA sequencing for the affected family members. RESULTS: Their phenotypes included severe symptoms in neonatal period, infantile onset of seizure and psychomotor delay and a mild late-onset form with speech delay at age 20months. All siblings were homozygous for a novel mutation Y164H in exon 4 of GLDC gene. The described novel homozygous variant in our study is predicted deleterious and pathogenic. CONCLUSIONS: This article further expands the genetic spectrum of glycine encephalopathy and adds an evidence of the clinical heterogeneity of glycine encephalopathy even in siblings with identical mutation.


Glycine Dehydrogenase (Decarboxylating)/genetics , Glycine Dehydrogenase (Decarboxylating)/metabolism , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism , Mutation , Arabs , Biomarkers/blood , Biomarkers/cerebrospinal fluid , Child , Child, Preschool , Female , Humans , Infant , Male , Siblings
6.
Ann Neurol ; 78(4): 606-18, 2015 Oct.
Article En | MEDLINE | ID: mdl-26179960

OBJECTIVE: Nonketotic hyperglycinemia is a neurometabolic disorder characterized by intellectual disability, seizures, and spasticity. Patients with attenuated nonketotic hyperglycinemia make variable developmental progress. Predictive factors have not been systematically assessed. METHODS: We reviewed 124 patients stratified by developmental outcome for biochemical and molecular predictive factors. Missense mutations were expressed to quantify residual activity using a new assay. RESULTS: Patients with severe nonketotic hyperglycinemia required multiple anticonvulsants, whereas patients with developmental quotient (DQ) > 30 did not require anticonvulsants. Brain malformations occurred mainly in patients with severe nonketotic hyperglycinemia (71%) but rarely in patients with attenuated nonketotic hyperglycinemia (7.5%). Neonatal presentation did not correlate with outcome, but age at onset ≥ 4 months was associated with attenuated nonketotic hyperglycinemia. Cerebrospinal fluid (CSF) glycine levels and CSF:plasma glycine ratio correlated inversely with DQ; CSF glycine > 230 µM indicated severe outcome and CSF:plasma glycine ratio ≤ 0.08 predicted attenuated outcome. The glycine index correlated strongly with outcome. Molecular analysis identified 99% of mutant alleles, including 96 novel mutations. Mutations near the active cleft of the P-protein maintained stable protein levels. Presence of 1 mutation with residual activity was necessary but not sufficient for attenuated outcome; 2 such mutations conferred best outcome. Divergent outcomes for the same genotype indicate a contribution of other genetic or nongenetic factors. INTERPRETATION: Accurate prediction of outcome is possible in most patients. A combination of 4 factors available neonatally predicted 78% of severe and 49% of attenuated patients, and a score based on mutation severity predicted outcome with 70% sensitivity and 97% specificity.


Glycine/genetics , Glycine/metabolism , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism , Mutation, Missense/genetics , Animals , COS Cells , Chlorocebus aethiops , Female , Glycine/chemistry , Humans , Hyperglycinemia, Nonketotic/diagnosis , Infant , Infant, Newborn , Male , Predictive Value of Tests , Prognosis , Protein Structure, Secondary
7.
Medicine (Baltimore) ; 93(7): e46, 2014 Aug.
Article En | MEDLINE | ID: mdl-25101986

Phenobarbital is an old antiepileptic drug used in severe epilepsy. Despite this, little is written about the need for dose adjustments in renal replacement therapy. Most sources recommend a moderately increased dose guided by therapeutic drug monitoring.A 14 year old boy with nonketotic hyperglycinemia, a rare inborn error of metabolism, characterized by high levels of glycine, epilepsy, spasticity, and cognitive impairment, was admitted to the emergency department with respiratory failure after a few days of fever and cough. The boy was unconscious at admittance and had acute renal and hepatic failure.Due to the acute respiratory infection, hypoxic hepatic and renal failure occurred and the patient had a status epilepticus.The patient was intubated and mechanically ventilated. Continuous renal replacement therapy was initiated. Despite increased phenobarbital doses, therapeutic levels were not reached until the dose was increased to 500 mg twice daily. Therapeutic drug monitoring was performed in plasma and dialysate. Calculations revealed that phenobarbital was almost freely dialyzed.Correct dosing of drugs in patients on renal replacement therapy may need a multidisciplinary approach and guidance by therapeutic drug monitoring.


Acute Kidney Injury/therapy , Anticonvulsants/pharmacokinetics , Hyperglycinemia, Nonketotic , Phenobarbital/pharmacokinetics , Renal Dialysis , Status Epilepticus/drug therapy , Acute Kidney Injury/etiology , Acute Kidney Injury/metabolism , Adolescent , Anticonvulsants/administration & dosage , Drug Monitoring/methods , Humans , Hyperglycinemia, Nonketotic/complications , Hyperglycinemia, Nonketotic/metabolism , Male , Phenobarbital/administration & dosage , Respiratory Syncytial Virus Infections/complications , Status Epilepticus/metabolism
8.
Metab Brain Dis ; 29(1): 211-3, 2014 Mar.
Article En | MEDLINE | ID: mdl-24407464

Early diagnosis for metabolic encephalopathy caused by inborn errors of metabolism is very important for the initiation of early treatment and also for prevention of sequela. Metabolic encephalopathy in the form of seizures can result from many inborn errors of metabolism and considering the large number of disorders causing metabolic encephalopathy, enzyme assays or conventional molecular tests are expensive and take considerably long period of time which results in delayed treatment. In our center we have used next generation DNA sequencing technology as an initial diagnostic test to look for about 700 disorders at the same time for the etiologic diagnosis of a 4-month-old female infant suffering from intractable seizures. The patient was found to have glycine encephalopathy resulting from a previously defined mutation in the GLDC gene. The diagnostic result was obtained much sooner than other conventional investigations. Up to our knowledge, this would be the first case with glycine encephalopathy in the literature who was approached by this novel panel method initially. Although currently, classical evaluation methods such as physical examination, biochemical and conventional molecular investigations are still accepted as the gold standards to clarify the etiology of the metabolic encephalopathy it is obvious that next generation sequence analysis will play a very significant role in the future.


Glycine Dehydrogenase (Decarboxylating)/genetics , Hyperglycinemia, Nonketotic/diagnosis , Mutation, Missense , Point Mutation , Sequence Analysis, DNA/methods , Base Sequence , DNA Mutational Analysis/methods , Female , Genes, Recessive , Glycine/blood , Glycine/cerebrospinal fluid , Glycine Dehydrogenase (Decarboxylating)/deficiency , Homozygote , Humans , Hyperglycinemia, Nonketotic/enzymology , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism , Infant , Molecular Sequence Data , Seizures/etiology , Sequence Alignment , Sequence Homology, Nucleic Acid
11.
Metab Brain Dis ; 27(3): 327-35, 2012 Sep.
Article En | MEDLINE | ID: mdl-22350964

Valproate administration increases the level of the inhibitory transmitter, glycine, in the urine and plasma of patients and experimental animals. Nonketotic hyperglycinemia (NKH), an autosomal recessive disorder of glycine metabolism, causes increased glycine concentrations in blood, urine, and cerebrospinal fluid (CSF), most likely due to a defect in the glycine cleavage enzyme or possibly deficits in glycine transport across cell membranes. We investigated the relationship between the hyperglycinemic effect of valproate and induced pyroglutamic aciduria via paracetamol in the vervet monkey. Firstly it was determined if valproate could induce hyperglycinemia in the monkey. The second aim was to increase glutamic acid (oxoproline) urine excretion using paracetamol as a pre-treatment and to assess whether valproate has an influence on the γ-glutamyl cycle. Hyperglycinemia was induced in healthy vervet monkeys when treated with a single oral dose of 50 mg/kg valproate. An acute dose of 50 mg/kg paracetamol increased oxoproline in the urine. Pre-treatment with paracetamol opposed the hyperglycinemic effect of valproate. However, the CSF:serum glycine ratio in a nonketotic monkey increased markedly after paracetamol treatment and remained high following valproate treatment. These results indicate that the γ-glutamyl cycle does indeed play a role in the hyperglycinemic effect of valproate treatment, and that paracetamol may have value in preventing and/or treating valproate-induced NKH.


Acetaminophen/pharmacology , Hyperglycinemia, Nonketotic/chemically induced , Hyperglycinemia, Nonketotic/prevention & control , Valproic Acid/toxicity , Acetaminophen/therapeutic use , Analgesics, Non-Narcotic/pharmacology , Analgesics, Non-Narcotic/therapeutic use , Animals , Anticonvulsants/toxicity , Chlorocebus aethiops , Disease Models, Animal , Female , Glycine/blood , Glycine/urine , Hyperglycinemia, Nonketotic/metabolism , Male , Pilot Projects
12.
Life Sci ; 89(7-8): 276-81, 2011 Aug 15.
Article En | MEDLINE | ID: mdl-21762704

AIMS: We investigated the effects of in vivo intrastriatal administration of glycine (Gly), which is found at high concentrations in the brain of patients affected by nonketotic hyperglycinemia (NKH), on important parameters of oxidative stress. MAIN METHODS: Thiobarbituric acid-reactive substances values (TBA-RS, lipid peroxidation), carbonyl formation (protein oxidative damage), sulfhydryl content, reduced glutathione concentrations, nitric oxide production and the activities of the antioxidant enzymes glutathione peroxidase, glutathione reductase, catalase, superoxide dismutase and glucose-6-phosphate dehydrogenase (antioxidant defenses) were measured in striatum from 30-day-old rats after Gly injection. KEY FINDINGS: Gly administration significantly increased TBA-RS values, implying lipid oxidative damage. Furthermore, Gly-induced increase of TBA-RS was fully prevented by the NMDA receptor antagonist MK-801, indicating the involvement of the NMDA glutamate receptor in this effect. Gly injection also induced protein carbonyl formation, as well as elevation of the activities of glutathione peroxidase, glutathione reductase, catalase and superoxide dismutase. In contrast, glutathione levels, sulfhydryl content, nitric oxide production and the activity of glucose-6-phosphate dehydrogenase were not modified by Gly. SIGNIFICANCE: The data shows that Gly in vivo administration causes lipid peroxidation, probably secondary to NMDA stimulation, induces protein oxidation and modulates the activities of important antioxidant enzymes in the striatum. In case these findings can be extrapolated to the human NKH, it is feasible that oxidative stress may be involved in the pathophysiology of the brain injury observed in patients with this neurometabolic disease.


Brain/drug effects , Corpus Striatum/drug effects , Glycine Agents/administration & dosage , Glycine/administration & dosage , Lipid Peroxidation/drug effects , Protein Carbonylation/drug effects , Animals , Brain/metabolism , Corpus Striatum/metabolism , Disease Models, Animal , Dizocilpine Maleate/pharmacology , Hyperglycinemia, Nonketotic/metabolism , Hyperglycinemia, Nonketotic/prevention & control , Microinjections , Neuroprotective Agents/pharmacology , Oxidoreductases/metabolism , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism , Thiobarbituric Acid Reactive Substances/metabolism
13.
Brain Dev ; 33(9): 753-7, 2011 Oct.
Article En | MEDLINE | ID: mdl-21470805

Glycine encephalopathy (GE), also known as non-ketotic hyperglycinemia, is a life-threatening metabolic disease caused by inherited deficiency of the glycine cleavage system (GCS). GE is characterized by accumulation of a large amount of glycine in serum and cerebrospinal fluids. In typical cases with GE, coma, profound hypotonia, and intractable seizures develop within several days of life. Patients with atypical symptoms may have delayed or missed diagnosis because of non-specific symptoms. It is sometimes problematic to confirm the diagnosis of GE since it requires either invasive liver biopsy for measurement of GCS activity or exhaustive mutational screening of three GCS genes, GLDC, AMT, and GCSH. We herein describe two novel laboratory tests for diagnosis of GE, [1-(13)C]glycine breath test and the multiplex ligation-dependent probe amplification (MLPA) for detection of large deletions in GLDC. The [1-(13)C]glycine breath test has been developed for noninvasive enzymatic diagnosis of GE. Because the GCS generates CO(2) by degradation of glycine, the GCS activity could be evaluated in vivo by measurement of exhaled (13)CO(2) after administration of a stable isotope, [1-(13)C]glycine. The MLPA has been developed for improvement in mutation detection rate in GE: Deletions involving multiple GDLC exons are prevalent among GE patients, but cannot be detected by the exon-sequencing analysis. Two novel diagnosis methods would facilitate diagnosis of hyperglycinemic patients as having GE.


Breath Tests/methods , Hyperglycinemia, Nonketotic/diagnosis , Multiplex Polymerase Chain Reaction/methods , Carbon Radioisotopes , Glycine/analysis , Glycine/genetics , Glycine Dehydrogenase (Decarboxylating)/genetics , Humans , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism
14.
J Comput Assist Tomogr ; 34(5): 762-5, 2010.
Article En | MEDLINE | ID: mdl-20861782

We present brain imaging and spectroscopy data in a neonate with a confirmed classic form of nonketotic hyperglycinemia (NKH), an autosomal-recessive metabolic disorder characterized by accumulation of glycine. To our knowledge, this is the first report of such complete analysis of the changes seen on conventional magnetic resonance imaging, diffusion-weighted imaging, and magnetic resonance spectroscopy at such an early age. The findings in a neonate are consistent with reports in older children with NKH, confirming that pathological changes typical for NKH can be seen in the first postnatal week.


Hyperglycinemia, Nonketotic/pathology , Anisotropy , Diffusion Magnetic Resonance Imaging , Female , Glycine/metabolism , Humans , Hyperglycinemia, Nonketotic/metabolism , Infant, Newborn , Magnetic Resonance Imaging , Magnetic Resonance Spectroscopy
15.
Cell Mol Neurobiol ; 29(2): 253-61, 2009 Mar.
Article En | MEDLINE | ID: mdl-18830815

Patients affected by nonketotic hyperglycinemia (NKH) usually present severe neurological symptoms and suffer from acute episodes of intractable seizures with leukoencephalopathy. Although excitotoxicity seems to be involved in the brain damage of NKH, the mechanisms underlying the neuropathology of this disease are not fully established. The objective of the present study was to investigate the in vitro effects of glycine (GLY), that accumulate at high concentrations in the brain of patients affected by this disorder, on important parameters of oxidative stress, such as lipid peroxidation (thiobarbituric acid-reactive substances (TBA-RS) and chemiluminescence) and the most important non-enzymatic antioxidant defense reduced glutathione (GSH) in cerebral cortex from 30-day-old rats. GLY significantly increased TBA-RS and chemiluminescence values, indicating that this metabolite provokes lipid oxidative damage. Furthermore, the addition of high doses of the antioxidants melatonin, trolox (soluble vitamin E) and GSH fully prevented GLY-induced increase of lipid peroxidation, indicating that free radicals were involved in this effect. GLY also decreased GSH brain concentrations, which was totally blocked by melatonin treatment. Finally, GLY significantly reduced sulfhydryl group content from a commercial GSH solution, but did not oxidize reduced cytochrome C. Our data indicate that oxidative stress elicited in vitro by GLY may possibly contribute at least in part to the pathophysiology of the neurological dysfunction in NKH.


Antioxidants/metabolism , Cerebral Cortex/metabolism , Glycine/metabolism , Hyperglycinemia, Nonketotic/metabolism , Lipid Peroxidation/physiology , Animals , Antioxidants/pharmacology , Cerebral Cortex/drug effects , Cerebral Cortex/physiopathology , Cytoprotection/drug effects , Cytoprotection/physiology , Dose-Response Relationship, Drug , Glutathione/metabolism , Glutathione/pharmacology , Glycine/toxicity , Hyperglycinemia, Nonketotic/physiopathology , Lipid Peroxidation/drug effects , Luminescence , Melatonin/metabolism , Melatonin/pharmacology , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Sulfhydryl Compounds/metabolism , Thiobarbituric Acid Reactive Substances/metabolism , Tocopherols/metabolism , Tocopherols/pharmacology
16.
Dev Med Child Neurol ; 50(2): 157-9, 2008 Feb.
Article En | MEDLINE | ID: mdl-18201306

Making a diagnosis of transient non-ketotic hyperglycinaemia (tNKH) can be difficult. We report an infant who presented in the neonatal period with symptoms of NKH. Metabolic studies performed on day 2 of life showed raised cerebrospinal fluid (CSF) and plasma glycine, and a CSF:plasma glycine ratio consistent with NKH; however, a liver biopsy performed on day 5 revealed normal liver glycine cleavage system activity. Subsequently, the child's clinical condition improved in the absence of any therapeutic medication. Clinical assessment and developmental follow-up at 5 months, 1 year, and 2 years were age-appropriate. Guidance for the investigation and management of future suspected cases of tNKH is discussed.


Glycine/metabolism , Hyperglycinemia, Nonketotic/diagnosis , Amino Acids/metabolism , Diagnosis, Differential , Female , Glycine/blood , Glycine/cerebrospinal fluid , Humans , Hyperglycinemia, Nonketotic/metabolism , Infant, Newborn , Liver/metabolism , Time Factors
17.
J Neuroendocrinol ; 19(12): 983-7, 2007 Dec.
Article En | MEDLINE | ID: mdl-18001328

Hypogonadotrophic hypogonadism is associated with uncontrolled diabetes mellitus. Hyperglycaemia is a unique metabolic abnormality of the hyperglycaemic hyperosmolar nonketotic state (HHNKS) and, as glucose availability regulates gonadotrophin release, we investigated whether gonadotrophin release is inhibited in diabetic women with HHNKS, and whether hyperglycaemia, hypernatraemia or both inhibit in vitro gonadotrophin-releasing hormone (GnRH) expression in GT1-7 neurones. Three groups of postmenopausal women were studied: nine diabetics with HHNKS, nine hospitalised ill nondiabetics and 15 healthy women. In addition, the effects of glucose (5.55, 33.3, 66.6 mmol/l) and sodium chloride (150 and 170 mmol/l) on GnRH expression were investigated using GT1-7 neurones. Postmenopausal diabetics with HHNKS showed a decrease in serum levels of luteinising hormone (diabetic HHNKS 2.2 +/- 0.9 IU/l versus ill nondiabetic 21.0 +/- 2.3 IU/l and healthy controls 20.9 +/- 2.8 IU/l, P < 0.01), follicle-stimulating hormone (diabetic HHNKS 8.2 +/- 2.1 IU/l versus ill nondiabetic 50.4 +/- 9.1 IU/l and controls 60.2 +/- 6.9 IU/l, P < 0.01) and free 3,5,3'-triiodothyronine (diabetic HHNKS 1.48 +/- 0.57 pmol/l versus ill nondiabetic 4.28 +/- 0.26 pmol/l and controls 3.88 +/- 0.11 pmol/l, P < 0.01). The plasma cortisol level was higher in both diabetic (985 +/- 130 nmol/l) and ill nondiabetic (726 +/- 52 nmol/l) women than in healthy women (512 +/- 47 nmol/l), but no differences were observed in plasma oestradiol, thyroid-stimulating hormone or free thyroxine. In vitro GT1-7 neurones expressed three-fold less GnRH at 170 mmol/l than at 150 mmol/l NaCl, whereas changing glucose concentrations in the culture medium did not affect GnRH expression. In conclusion, postmenopausal diabetic women with HHNKS show decreased serum gonadotrophin levels, and severe hypernatraemia may participate in the hypogonadotropism observed in HHNKS.


Diabetes Mellitus/metabolism , Gonadotropins/deficiency , Hyperglycinemia, Nonketotic/metabolism , Hypernatremia/metabolism , Postmenopause/metabolism , Aged , Aged, 80 and over , Blood Glucose/metabolism , Diabetes Mellitus/blood , Female , Gonadotropin-Releasing Hormone/metabolism , Gonadotropins/blood , Hormones/blood , Humans , Middle Aged , Postmenopause/blood , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction
18.
J Child Neurol ; 21(10): 900-3, 2006 Oct.
Article En | MEDLINE | ID: mdl-17005111

Neonatal nonketotic hyperglycinemia is usually fatal or, less commonly, severely developmentally disabling, whereas transient nonketotic hyperglycinemia has usually been followed by normal development. We report a boy who had transient neonatal nonketotic hyperglycinemia but a coexistent disorder of serotonin metabolism manifested by initially low cerebrospinal fluid 5-hydroxyindoleacetic acid (which later normalized), low whole blood serotonin, and decreased platelet serotonin uptake. He survived the neonatal period but was neurodevelopmentally delayed and developed an autistic-like disorder. Later, his positron emission tomographic (PET) scans with alpha[(11)C] methyl-l-tryptophan revealed a pattern characteristic of autistic children. Although we know of no link between glycine and serotonin metabolism, and our patient had low, rather than high, central and peripheral serotonin, this case might represent a novel infantile disorder that affects both the glycine and serotonin neurotransmitter systems.


Hyperglycinemia, Nonketotic/complications , Hyperglycinemia, Nonketotic/metabolism , Seizures/complications , Seizures/metabolism , Serotonin/deficiency , Brain Diseases, Metabolic/complications , Child , Follow-Up Studies , Humans , Hyperglycinemia, Nonketotic/pathology , Male , Seizures/pathology
19.
Handb Exp Pharmacol ; (175): 457-83, 2006.
Article En | MEDLINE | ID: mdl-16722246

Glycine has multiple neurotransmitter functions in the central nervous system (CNS). In the spinal cord and brainstem of vertebrates, it serves as a major inhibitory neurotransmitter. In addition, it participates in excitatory neurotransmission by modulating the activity of the N-methyl-D-aspartate (NMDA) subtype of glutamate receptors. The extracellular concentrations of glycine are regulated by Na+/Cl(-)-dependent glycine transporters (GlyTs), which are expressed in neurons and adjacent glial cells. Considerable progress has been made recently towards elucidating the in vivo roles of GlyTs in the CNS. The generation and analysis of animals carrying targeted disruptions of GlyT genes (GlyT knockout mice) have allowed investigators to examine the different contributions of individual GlyT subtypes to synaptic transmission. In addition, they have provided animal models for two hereditary human diseases, glycine encephalopathy and hyperekplexia. Selective GlyT inhibitors have been shown to modulate neurotransmission and might constitute promising therapeutic tools for the treatment of psychiatric and neurological disorders such as schizophrenia and pain. Therefore, pharmacological and genetic studies indicate that GlyTs are key regulators of both glycinergic inhibitory and glutamatergic excitatory neurotransmission. This chapter describes our present understanding of the functions of GlyTs and their involvement in the fine-tuning of neuronal communication.


Glycine Plasma Membrane Transport Proteins/metabolism , Glycine/metabolism , Synaptic Transmission , Animals , Brain/metabolism , Disease Models, Animal , Gene Expression Regulation , Glycine Plasma Membrane Transport Proteins/deficiency , Glycine Plasma Membrane Transport Proteins/genetics , Humans , Hyperglycinemia, Nonketotic/genetics , Hyperglycinemia, Nonketotic/metabolism , Mice , Mice, Knockout , Receptors, N-Methyl-D-Aspartate/metabolism , Reflex, Abnormal/genetics , Transcription, Genetic
20.
J Mol Biol ; 351(5): 1146-59, 2005 Sep 02.
Article En | MEDLINE | ID: mdl-16051266

T-protein, a component of the glycine cleavage system, catalyzes the formation of ammonia and 5,10-methylenetetrahydrofolate from the aminomethyl moiety of glycine attached to the lipoate cofactor of H-protein. Several mutations in the human T-protein gene cause non-ketotic hyperglycinemia. To gain insights into the effect of disease-causing mutations and the catalytic mechanism at the molecular level, crystal structures of human T-protein in free form and that bound to 5-methyltetrahydrofolate (5-CH3-H4folate) have been determined at 2.0 A and 2.6 A resolution, respectively. The overall structure consists of three domains arranged in a cloverleaf-like structure with the central cavity, where 5-CH3-H4folate is bound in a kinked shape with the pteridine group deeply buried into the hydrophobic pocket and the glutamyl group pointed to the C-terminal side surface. Most of the disease-related residues cluster around the cavity, forming extensive hydrogen bonding networks. These hydrogen bonding networks are employed in holding not only the folate-binding space but also the positions and the orientations of alpha-helix G and the following loop in the middle region, which seems to play a pivotal role in the T-protein catalysis. Structural and mutational analyses demonstrated that Arg292 interacts through water molecules with the folate polyglutamate tail, and that the invariant Asp101, located close to the N10 group of 5-CH3-H4folate, might play a key role in the initiation of the catalysis by increasing the nucleophilic character of the N10 atom of the folate substrate for the nucleophilic attack on the aminomethyl lipoate intermediate. A clever mechanism of recruiting the aminomethyl lipoate arm to the reaction site seems to function as a way of avoiding the release of toxic formaldehyde.


Crystallography, X-Ray/methods , Glycine/chemistry , Hydroxymethyl and Formyl Transferases/chemistry , Hyperglycinemia, Nonketotic/metabolism , Amino Acid Sequence , Aminomethyltransferase , Animals , Arginine/chemistry , Asparagine/chemistry , Binding Sites , Catalytic Domain , Cluster Analysis , DNA Mutational Analysis , Formaldehyde/chemistry , Humans , Hydrogen Bonding , Kinetics , Models, Chemical , Models, Molecular , Molecular Conformation , Molecular Sequence Data , Mutation , Protein Binding , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Homology, Amino Acid
...