Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 99
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Hum Mol Genet ; 32(6): 917-933, 2023 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-36190515

RESUMO

Maintaining protein lipoylation is vital for cell metabolism. The H-protein encoded by GCSH has a dual role in protein lipoylation required for bioenergetic enzymes including pyruvate dehydrogenase and 2-ketoglutarate dehydrogenase, and in the one-carbon metabolism through its involvement in glycine cleavage enzyme system, intersecting two vital roles for cell survival. Here, we report six patients with biallelic pathogenic variants in GCSH and a broad clinical spectrum ranging from neonatal fatal glycine encephalopathy to an attenuated phenotype of developmental delay, behavioral problems, limited epilepsy and variable movement problems. The mutational spectrum includes one insertion c.293-2_293-1insT, one deletion c.122_(228 + 1_229-1) del, one duplication of exons 4 and 5, one nonsense variant p.Gln76*and four missense p.His57Arg, p.Pro115Leu and p.Thr148Pro and the previously described p.Met1?. Via functional studies in patient's fibroblasts, molecular modeling, expression analysis in GCSH knockdown COS7 cells and yeast, and in vitro protein studies, we demonstrate for the first time that most variants identified in our cohort produced a hypomorphic effect on both mitochondrial activities, protein lipoylation and glycine metabolism, causing combined deficiency, whereas some missense variants affect primarily one function only. The clinical features of the patients reflect the impact of the GCSH changes on any of the two functions analyzed. Our analysis illustrates the complex interplay of functional and clinical impact when pathogenic variants affect a multifunctional protein involved in two metabolic pathways and emphasizes the value of the functional assays to select the treatment and investigate new personalized options.


Assuntos
Hiperglicinemia não Cetótica , Humanos , Hiperglicinemia não Cetótica/genética , Hiperglicinemia não Cetótica/patologia , Proteínas/genética , Mutação , Éxons/genética , Glicina/genética , Glicina/metabolismo
2.
Mol Genet Metab ; 142(3): 108496, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38761651

RESUMO

Non-Ketotic Hyperglycinemia (NKH) is a rare inborn error of metabolism caused by impaired function of the glycine cleavage system (GCS) and characterised by accumulation of glycine in body fluids and tissues. NKH is an autosomal recessive condition and the majority of affected individuals carry mutations in GLDC (glycine decarboxylase). Current treatments for NKH have limited effect and are not curative. As a monogenic condition with known genetic causation, NKH is potentially amenable to gene therapy. An AAV9-based expression vector was designed to target sites of GCS activity. Using a ubiquitous promoter to drive expression of a GFP reporter, transduction of liver and brain was confirmed following intra-venous and/or intra-cerebroventricular administration to neonatal mice. Using the same capsid and promoter with transgenes to express mouse or human GLDC, vectors were then tested in GLDC-deficient mice that provide a model of NKH. GLDC-deficient mice exhibited elevated plasma glycine concentration and accumulation of glycine in liver and brain tissues as previously observed. Moreover, the folate profile indicated suppression of folate one­carbon metabolism (FOCM) in brain tissue, as found at embryonic stages, and reduced abundance of FOCM metabolites including betaine and choline. Neonatal administration of vector achieved reinstatement of GLDC mRNA and protein expression in GLDC-deficient mice. Treated GLDC-deficient mice showed significant lowering of plasma glycine, confirming functionality of vector expressed protein. AAV9-GLDC treatment also led to lowering of brain tissue glycine, and normalisation of the folate profile indicating restoration of glycine-derived one­carbon supply. These findings support the hypothesis that AAV-mediated gene therapy may offer potential in treatment of NKH.


Assuntos
Encéfalo , Dependovirus , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos , Glicina Desidrogenase (Descarboxilante) , Glicina , Hiperglicinemia não Cetótica , Fígado , Animais , Hiperglicinemia não Cetótica/genética , Hiperglicinemia não Cetótica/metabolismo , Hiperglicinemia não Cetótica/terapia , Glicina Desidrogenase (Descarboxilante)/genética , Glicina Desidrogenase (Descarboxilante)/metabolismo , Dependovirus/genética , Camundongos , Humanos , Vetores Genéticos/genética , Glicina/metabolismo , Fígado/metabolismo , Encéfalo/metabolismo , Biomarcadores/metabolismo , Ácido Fólico/metabolismo
3.
Am J Med Genet A ; 194(8): e63622, 2024 08.
Artigo em Inglês | MEDLINE | ID: mdl-38572626

RESUMO

Nonketotic hyperglycinemia (NKH) is a relatively well-characterized inborn error of metabolism that results in a combination of lethargy, hypotonia, seizures, developmental arrest, and, in severe cases, death early in life. Three genes encoding components of the glycine cleavage enzyme system-GLDC, AMT, and GCSH-are independently associated with NKH. We report on a patient with severe NKH in whom the homozygous pathogenic variant in AMT (NM_000481.3):c.602_603del (p.Lys201Thrfs*75) and the homozygous likely pathogenic variant in GLDC(NM_000170.2):c.2852C>A (p.Ser951Tyr) were both identified. Our patient demonstrates a novel combination of two homozygous disease-causing variants impacting the glycine cleavage pathway at two different components, and elicits management- and genetic counseling-related challenges for the family.


Assuntos
Homozigoto , Hiperglicinemia não Cetótica , Humanos , Hiperglicinemia não Cetótica/genética , Hiperglicinemia não Cetótica/patologia , Masculino , Glicina Desidrogenase (Descarboxilante)/genética , Aminometiltransferase/genética , Feminino , Mutação/genética , Lactente , Glicina/genética , Recém-Nascido , Fenótipo , Predisposição Genética para Doença , Aminoácido Oxirredutases , Complexos Multienzimáticos , Transferases
4.
PLoS Genet ; 17(2): e1009307, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33524012

RESUMO

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.


Assuntos
Modelos Animais de Doenças , Glicina Desidrogenase (Descarboxilante)/química , Glicina Desidrogenase (Descarboxilante)/genética , Glicina/metabolismo , Hiperglicinemia não Cetótica/genética , Animais , Feminino , Genômica , Genótipo , Glicina/genética , Humanos , Hiperglicinemia não Cetótica/metabolismo , Hiperglicinemia não Cetótica/patologia , Masculino , Espectrometria de Massas , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mutação , Mutação de Sentido Incorreto , Fenótipo
5.
Int J Mol Sci ; 25(5)2024 Feb 28.
Artigo em Inglês | MEDLINE | ID: mdl-38474060

RESUMO

The pathophysiology of nonketotic hyperglycinemia (NKH), a rare neuro-metabolic disorder associated with severe brain malformations and life-threatening neurological manifestations, remains incompletely understood. Therefore, a valid human neural model is essential. We aimed to investigate the impact of GLDC gene variants, which cause NKH, on cellular fitness during the differentiation process of human induced pluripotent stem cells (iPSCs) into iPSC-derived astrocytes and to identify sustainable mechanisms capable of overcoming GLDC deficiency. We developed the GLDC27-FiPS4F-1 line and performed metabolomic, mRNA abundance, and protein analyses. This study showed that although GLDC27-FiPS4F-1 maintained the parental genetic profile, it underwent a metabolic switch to an altered serine-glycine-one-carbon metabolism with a coordinated cell growth and cell cycle proliferation response. We then differentiated the iPSCs into neural progenitor cells (NPCs) and astrocyte-lineage cells. Our analysis showed that GLDC-deficient NPCs had shifted towards a more heterogeneous astrocyte lineage with increased expression of the radial glial markers GFAP and GLAST and the neuronal markers MAP2 and NeuN. In addition, we detected changes in other genes related to serine and glycine metabolism and transport, all consistent with the need to maintain glycine at physiological levels. These findings improve our understanding of the pathology of nonketotic hyperglycinemia and offer new perspectives for therapeutic options.


Assuntos
Hiperglicinemia não Cetótica , Células-Tronco Pluripotentes Induzidas , Humanos , Hiperglicinemia não Cetótica/genética , Hiperglicinemia não Cetótica/patologia , Glicina Desidrogenase (Descarboxilante)/genética , Astrócitos/patologia , Células-Tronco Pluripotentes Induzidas/patologia , Glicina , Serina
6.
Ann Neurol ; 92(2): 292-303, 2022 08.
Artigo em Inglês | MEDLINE | ID: mdl-35616651

RESUMO

OBJECTIVE: Glycine encephalopathy, also known as nonketotic hyperglycinemia (NKH), is an inherited neurometabolic disorder with variable clinical course and severity, ranging from infantile epileptic encephalopathy to psychiatric disorders. A precise phenotypic characterization and an evaluation of predictive approaches are needed. METHODS: Longitudinal clinical and biochemical data of 25 individuals with NKH from the patient registry of the International Working Group on Neurotransmitter Related Disorders were studied with in silico analyses, pathogenicity scores, and molecular modeling of GLDC and AMT variants. RESULTS: Symptom onset (p < 0.01) and diagnosis occur earlier in life in severe NKH (p < 0.01). Presenting symptoms affect the age at diagnosis. Psychiatric problems occur predominantly in attenuated NKH. Onset age ≥ 3 months (66% specificity, 100% sensitivity, area under the curve [AUC] = 0.87) and cerebrospinal fluid (CSF)/plasma glycine ratio ≤ 0.09 (57% specificity, 100% sensitivity, AUC = 0.88) are sensitive indicators for attenuated NKH, whereas CSF glycine concentration ≥ 116.5µmol/l (100% specificity, 93% sensitivity, AUC = 0.97) and CSF/plasma glycine ratio ≥ 0.15 (100% specificity, 64% sensitivity, AUC = 0.88) are specific for severe forms. A ratio threshold of 0.128 discriminates the overlapping range. We present 10 new GLDC variants. Two mild variants resulted in attenuated, whereas 2 severe variants or 1 mild and 1 severe variant led to severe phenotype. Based on clinical, biochemical, and genetic parameters, we propose a severity prediction model. INTERPRETATION: This study widens the phenotypic spectrum of attenuated NKH and expands the number of pathogenic variants. The multiparametric approach provides a promising tool to predict disease severity, helping to improve clinical management strategies. ANN NEUROL 2022;92:292-303.


Assuntos
Hiperglicinemia não Cetótica , Glicina/líquido cefalorraquidiano , Glicina/genética , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Hiperglicinemia não Cetótica/genética , Hiperglicinemia não Cetótica/patologia , Mutação , Fenótipo
7.
Clin Genet ; 100(2): 201-205, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33890291

RESUMO

The glycine cleavage system H protein (GCSH) is an integral part of the glycine cleavage system with its additional involvement in the synthesis and transport of lipoic acid. We hypothesize that pathogenic variants in GCSH can cause variant nonketotic hyperglycinemia (NKH), a heterogeneous group of disorders with findings resembling a combination of severe NKH (elevated levels of glycine in plasma and CSF, progressive lethargy, seizures, severe hypotonia, no developmental progress, early death) and mitochondriopathies (lactic acidosis, leukoencephalopathy and Leigh-like lesions on MRI). We herein report three individuals from two unrelated Indian families with clinical, biochemical, and radiological findings of variant NKH, harboring a biallelic start loss variant, c.1A > G in GCSH.


Assuntos
Proteína H do Complexo Glicina Descarboxilase/genética , Hiperglicinemia não Cetótica/genética , Pré-Escolar , Feminino , Glicina/sangue , Glicina/líquido cefalorraquidiano , Humanos , Hiperglicinemia não Cetótica/etiologia , Masculino , Mutação , Linhagem
8.
Am J Med Genet A ; 185(2): 476-485, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33269555

RESUMO

GLYT1 encephalopathy is a form of glycine encephalopathy caused by disturbance of glycine transport. The phenotypic spectrum of the disease has not yet been completely described, as only four unrelated families with the disorder have been reported to date. Common features of affected patients include neonatal hypotonia, respiratory failure, encephalopathy, myoclonic jerks, dysmorphic features, and musculoeskeletal anomalies. All reported affected patients harbor biallelic genetic variants in SLC6A9. SNP array together with Sanger sequencing were performed in a newborn with arthrogryposis and severe neurological impairment. The novel genetic variant c.997delC in SLC6A9 was detected in homozygous state in the patient. At protein level, the predicted change is p.(Arg333Alafs*3), which most probably results in a loss of protein function. The variant cosegregated with the disease in the family. A subsequent pregnancy with ultrasound anomalies was also affected. The proband presented the core phenotypic features of GLYT1 encephalopathy, but also a burst suppression pattern on the electroencephalogram, a clinical feature not previously associated with the disorder. Our results suggest that the appearance of this pattern correlates with higher cerebrospinal fluid glycine levels and cerebrospinal fluid/plasma glycine ratios. A detailed discussion on the possible pathophysiological mechanisms of the disorder is also provided.


Assuntos
Artrogripose/genética , Predisposição Genética para Doença , Proteínas da Membrana Plasmática de Transporte de Glicina/genética , Hiperglicinemia não Cetótica/genética , Aborto Espontâneo/genética , Aborto Espontâneo/patologia , Artrogripose/mortalidade , Artrogripose/patologia , Feminino , Glicina/genética , Glicina/metabolismo , Homozigoto , Humanos , Hiperglicinemia não Cetótica/mortalidade , Hiperglicinemia não Cetótica/patologia , Recém-Nascido , Masculino , Mutação/genética , Linhagem , Fenótipo
9.
PLoS Comput Biol ; 16(5): e1007871, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32421718

RESUMO

Monogenetic diseases provide unique opportunity for studying complex, clinical states that underlie neurological severity. Loss of glycine decarboxylase (GLDC) can severely impact neurological development as seen in non-ketotic hyperglycinemia (NKH). NKH is a neuro-metabolic disorder lacking quantitative predictors of disease states. It is characterized by elevation of glycine, seizures and failure to thrive, but glycine reduction often fails to confer neurological benefit, suggesting need for alternate tools to distinguish severe from attenuated disease. A major challenge has been that there are 255 unique disease-causing missense mutations in GLDC, of which 206 remain entirely uncharacterized. Here we report a Multiparametric Mutation Score (MMS) developed by combining in silico predictions of stability, evolutionary conservation and protein interaction models and suitable to assess 251 of 255 mutations. In addition, we created a quantitative scale of clinical disease severity comprising of four major disease domains (seizure, cognitive failure, muscular and motor control and brain-malformation) to comprehensively score patient symptoms identified in 131 clinical reports published over the last 15 years. The resulting patient Clinical Outcomes Scores (COS) were used to optimize the MMS for biological and clinical relevance and yield a patient Weighted Multiparametric Mutation Score (WMMS) that separates severe from attenuated neurological disease (p = 1.2 e-5). Our study provides understanding for developing quantitative tools to predict clinical severity of neurological disease and a clinical scale that advances monitoring disease progression needed to evaluate new treatments for NKH.


Assuntos
Regulação Enzimológica da Expressão Gênica , Genótipo , Glicina Desidrogenase (Descarboxilante)/genética , Hiperglicinemia não Cetótica/genética , Mutação de Sentido Incorreto , Fenótipo , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Hiperglicinemia não Cetótica/patologia , Índice de Gravidade de Doença
10.
Metab Brain Dis ; 36(6): 1213-1222, 2021 08.
Artigo em Inglês | MEDLINE | ID: mdl-33791923

RESUMO

Nonketotic hyperglycinemia is an autosomal recessive inborn error of glycine metabolism, characterized by deficient activity of the glycine cleavage enzyme system. Classic nonketotic hyperglycinemia is caused by mutations or genomic changes in genes that encode the protein components of the glycine cleavage enzyme system. We aimed to investigate clinical, biochemical, radiological findings and molecular genetic data in ten Turkish patients with classic nonketotic hyperglycinemia. Ten Turkish patients who were diagnosed with classic nonketotic hyperglycinemia in a single center from 2013 to 2019 were included in this study. Their clinical, radiological, electrophysiological and laboratory data were collected retrospectively. Sixty percent of the patients were in neonatal group, while 40 % of the patients were infantile. There were no late-onset patients. 90 % of the patients had the severe form. All patients had developmental delay and seizures. Mortality ratio was 30 % in all groups and 50 % in the neonatal group, while no mortality was seen in infantile group. Median (range) values of cerebrospinal fluid (CSF) glycine levels, plasma glycine levels and CSF/plasma glycine ratios were 148 (15-320) µmol/L, 896 (87-1910) µmol/L, 0.17 (0.09-0.21) respectively. Diffuse hypomyelination and corpus callosum anomaly were the most common cranial MRI findings and multifocal epileptic activity and burst supression pattern were the most common electroencephalographic findings. Six patients had variants in GLDC gene and four in AMT gene; five novel variants including AMT gene deletion were detected. Prognosis was poor and treatment was not effective, especially in the severe form. Classic nonketotic hyperglycinemia causes high morbidity and mortality. Neonatal-onset disease was more common and severe than infantile-onset disease. The ratio of AMT gene variants might be higher in Turkey than other countries. AMT gene deletion also plays a role in the etiology of classic nonketotic hyperglycinemia.


Assuntos
Genótipo , Hiperglicinemia não Cetótica/genética , Mutação/genética , Convulsões/etiologia , Agenesia do Corpo Caloso , Aminoácido Oxirredutases/genética , Feminino , Glicina/metabolismo , Humanos , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Masculino , Complexos Multienzimáticos/genética , Estudos Retrospectivos , Convulsões/genética , Transferases/genética
11.
Am J Hum Genet ; 99(5): 1172-1180, 2016 Nov 03.
Artigo em Inglês | MEDLINE | ID: mdl-27773429

RESUMO

Glycine is a major neurotransmitter that activates inhibitory glycine receptors and is a co-agonist for excitatory glutamatergic N-methyl-D-aspartate (NMDA) receptors. Two transporters, GLYT1 and GLYT2, regulate extracellular glycine concentrations within the CNS. Dysregulation of the extracellular glycine has been associated with hyperekplexia and nonketotic hyperglycinemia. Here, we report four individuals from two families who presented at birth with facial dysmorphism, encephalopathy, arthrogryposis, hypotonia progressing to hypertonicity with startle-like clonus, and respiratory failure. Only one individual survived the respiratory failure and was weaned off ventilation but has significant global developmental delay. Mildly elevated cerebrospinal fluid (CSF) glycine and normal serum glycine were observed in two individuals. In both families, we identified truncating mutations in SLC6A9, encoding GLYT1. We demonstrate that pharmacologic or genetic abolishment of GlyT1 activity in mice leads to mildly elevated glycine in the CSF but not in blood. Additionally, previously reported slc6a9-null mice and zebrafish mutants also display phenotypes consistent with the affected individuals we examined. Our data suggest that truncating SLC6A9 mutations lead to a distinct human neurological syndrome hallmarked by mildly elevated CSF glycine and normal serum glycine.


Assuntos
Artrogripose/genética , Proteínas da Membrana Plasmática de Transporte de Glicina/genética , Glicina/líquido cefalorraquidiano , Hiperglicinemia não Cetótica/genética , Animais , Artrogripose/diagnóstico , Pré-Escolar , Feminino , Deleção de Genes , Regulação da Expressão Gênica , Glicina/sangue , Proteínas da Membrana Plasmática de Transporte de Glicina/metabolismo , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Lactente , Recém-Nascido , Masculino , Camundongos , Camundongos Knockout , Linhagem
12.
J Med Primatol ; 48(3): 161-165, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30724368

RESUMO

BACKGROUND: Nonketotic hyperglycinemia (NKH) is a rare metabolic disorder that is characterized by high levels of glycine in plasma and cerebrospinal fluid in humans. In this study, total congenital cataract captive-bred Vervet monkeys (Chlorocebus aethiops) that are hyperglycinemic were screened to identify mutations in Bola type 3 (BOLA3), glutaredoxin 5 (GLRX5), and lipoate synthase (LIAS) genes. METHODS: Twenty-four Vervet monkeys (12 hyperglycinemic and 12 healthy controls) were selected for mutation analysis using polymerase chain reaction (PCR), Sanger sequencing, and reverse transcriptase-polymerase chain reaction (RT-PCR). RESULTS: Novel sequence variants were identified in BOLA3 (R23H and Q38R) and LIAS (R369I and A371A), and gene expression in the control group was significantly lower compared to the hyperglycinemic group (P < 0.05). CONCLUSION: The data obtained from this study will contribute to generation of new knowledge regarding the involvement of these genes in NKH development.


Assuntos
Catarata/congênito , Chlorocebus aethiops , Doenças Genéticas Ligadas ao Cromossomo X/veterinária , Hiperglicinemia não Cetótica/veterinária , Microftalmia/veterinária , Doenças dos Macacos/genética , Animais , Animais de Zoológico , Catarata/genética , Catarata/veterinária , Doenças Genéticas Ligadas ao Cromossomo X/genética , Hiperglicinemia não Cetótica/genética , Microftalmia/genética
13.
Metab Brain Dis ; 34(1): 373-376, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30350008

RESUMO

Nonketotic Hyperglycinemia is an autosomal recessive disorder characterized by defects in the mitochondrial glycine cleavage system. Most patients present soon after birth with seizures and hypotonia, and infants that survive the newborn period often have profound intellectual disability and intractable seizures. Here we present a case report of a 4-year-old girl with NKH as well as hyperammonemia, an uncommon finding in NKH. Genetic analysis found a previously unreported homozygous mutation (c.878-1 G > A) in the AMT gene. Maximum Entropy Principle modeling predicted that this mutation most likely breaks the splice site at the border of intron 7 and exon 8 of the AMT gene. Treatment with L-Arginine significantly reduced both the proband's glycine and ammonia levels, in turn aiding in control of seizures and mental status. This is the first time the use of L-Arginine is reported to successfully treat elevated glycine levels.


Assuntos
Aminometiltransferase/genética , Hiperamonemia/genética , Hiperglicinemia não Cetótica/genética , Íntrons , Mutação , Pré-Escolar , Feminino , Homozigoto , Humanos , Hiperamonemia/complicações , Hiperglicinemia não Cetótica/complicações
14.
BMC Med Genet ; 19(1): 5, 2018 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-29304759

RESUMO

BACKGROUND: Non-ketotic hyperglycinemia (NKH) is a rare, devastating autosomal recessive disorder of glycine metabolism with a very poor prognosis. Currently, few studies have reported genetic profiling of Chinese NKH patients. This study aimed to identify the genetic mutations in a Chinese family with NKH. METHODS: A Chinese family of Han ethnicity, with three siblings with NKH was studied. Sanger sequencing and multiplex ligation-dependent probe amplification combined with SYBR green real-time quantitative PCR was used to identify potential mutations in the GLDC, AMT and GCSH genes. The potential pathogenicity of the identified missense mutation was analyzed using SIFT, PolyPhen-2, PROVEAN and MutationTaster software. RESULTS: All patients exhibited severe and progressive clinical symptoms, including lethargy, hypotonia and seizures, and had greatly elevated glycine levels in their plasma and CSF. Molecular genetic analysis identified compound heterozygous variants in the GLDC gene in these three siblings, including a novel missense variant c.2680A > G (p.Thr894Ala) in exon 23 and a heterozygous deletion of exon 3, which were inherited respectively from their parents. In silico analysis, using several different types of bioinformatic software, predicted that the novel variant c.2680A > G in the GLDC gene was pathogenic. Moreover, the deletion of exon 3 was identified for the first time in a Chinese population. CONCLUSIONS: A novel missense variant and a previously reported deletion in GLDC gene were identified. The two variants of GLDC gene identified probably underlie the pathogenesis of non-ketotic hyperglycinemia in this family, and also enrich the mutational spectrum of GLDC gene.


Assuntos
Povo Asiático/genética , Glicina Desidrogenase (Descarboxilante)/genética , Heterozigoto , Hiperglicinemia não Cetótica/genética , Sequência de Aminoácidos , China , Éxons , Feminino , Variação Genética , Genótipo , Glicina/sangue , Glicina/líquido cefalorraquidiano , Humanos , Hiperglicinemia não Cetótica/enzimologia , Recém-Nascido , Masculino , Mutação de Sentido Incorreto , Linhagem , Deleção de Sequência , Irmãos
15.
J Theor Biol ; 455: 97-100, 2018 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-30009793

RESUMO

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.


Assuntos
Encéfalo/metabolismo , Glicina/metabolismo , Hiperglicinemia não Cetótica/metabolismo , Modelos Neurológicos , Doenças do Sistema Nervoso/metabolismo , Células-Tronco Neurais/metabolismo , Encéfalo/anormalidades , Encéfalo/patologia , Proliferação de Células , DNA/genética , Glicina/genética , Humanos , Hiperglicinemia não Cetótica/genética , Hiperglicinemia não Cetótica/patologia , Doenças do Sistema Nervoso/genética , Doenças do Sistema Nervoso/patologia , Células-Tronco Neurais/patologia
16.
Hum Mutat ; 38(6): 678-691, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28244183

RESUMO

The rapid analysis of genomic data is providing effective mutational confirmation in patients with clinical and biochemical hallmarks of a specific disease. This is the case for nonketotic hyperglycinemia (NKH), a Mendelian disorder causing seizures in neonates and early-infants, primarily due to mutations in the GLDC gene. However, understanding the impact of missense variants identified in this gene is a major challenge for the application of genomics into clinical practice. Herein, a comprehensive functional and structural analysis of 19 GLDC missense variants identified in a cohort of 26 NKH patients was performed. Mutant cDNA constructs were expressed in COS7 cells followed by enzymatic assays and Western blot analysis of the GCS P-protein to assess the residual activity and mutant protein stability. Structural analysis, based on molecular modeling of the 3D structure of GCS P-protein, was also performed. We identify hypomorphic variants that produce attenuated phenotypes with improved prognosis of the disease. Structural analysis allows us to interpret the effects of mutations on protein stability and catalytic activity, providing molecular evidence for clinical outcome and disease severity. Moreover, we identify an important number of mutants whose loss-of-functionality is associated with instability and, thus, are potential targets for rescue using folding therapeutic approaches.


Assuntos
Glicina Desidrogenase (Descarboxilante)/genética , Hiperglicinemia não Cetótica/genética , Mutação de Sentido Incorreto/genética , Relação Estrutura-Atividade , Éxons/genética , Regulação Enzimológica da Expressão Gênica , Glicina/metabolismo , Glicina Desidrogenase (Descarboxilante)/química , Humanos , Hiperglicinemia não Cetótica/patologia , Recém-Nascido , Conformação Molecular , Fenótipo , Estabilidade Proteica
17.
Genet Med ; 19(1): 104-111, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27362913

RESUMO

PURPOSE: The study's purpose was to delineate the genetic mutations that cause classic nonketotic hyperglycinemia (NKH). METHODS: Genetic results, parental phase, ethnic origin, and gender data were collected from subjects suspected to have classic NKH. Mutations were compared with those in the existing literature and to the population frequency from the Exome Aggregation Consortium (ExAC) database. RESULTS: In 578 families, genetic analyses identified 410 unique mutations, including 246 novel mutations. 80% of subjects had mutations in GLDC. Missense mutations were noted in 52% of all GLDC alleles, most private. Missense mutations were 1.5 times as likely to be pathogenic in the carboxy terminal of GLDC than in the amino-terminal part. Intragenic copy-number variations (CNVs) in GLDC were noted in 140 subjects, with biallelic CNVs present in 39 subjects. The position and frequency of the breakpoint for CNVs correlated with intron size and presence of Alu elements. Missense mutations, most often recurring, were the most common type of disease-causing mutation in AMT. Sequencing and CNV analysis identified biallelic pathogenic mutations in 98% of subjects. Based on genotype, 15% of subjects had an attenuated phenotype. The frequency of NKH is estimated at 1:76,000. CONCLUSION: The 484 unique mutations now known in classic NKH provide a valuable overview for the development of genotype-based therapies.Genet Med 19 1, 104-111.


Assuntos
Aminometiltransferase/genética , Complexo Glicina Descarboxilase/genética , Glicina Desidrogenase (Descarboxilante)/genética , Hiperglicinemia não Cetótica/genética , Alelos , Di-Hidrolipoamida Desidrogenase/genética , Éxons/genética , Feminino , Testes Genéticos , Genótipo , Glicina/genética , Glicina/metabolismo , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Hiperglicinemia não Cetótica/patologia , Íntrons , Masculino , Mutação de Sentido Incorreto
18.
Mol Genet Metab ; 121(2): 80-82, 2017 06.
Artigo em Inglês | MEDLINE | ID: mdl-28462797

RESUMO

Historically, d-glyceric aciduria was thought to cause an uncharacterized blockage to the glycine cleavage enzyme system (GCS) causing nonketotic hyperglycinemia (NKH) as a secondary phenomenon. This inference was reached based on the clinical and biochemical results from the first d-glyceric aciduria patient reported in 1974. Along with elevated glyceric acid excretion, this patient exhibited severe neurological symptoms of myoclonic epilepsy and absent development, and had elevated glycine levels and decreased glycine cleavage system enzyme activity. Mutations in the GLYCTK gene (encoding d-glycerate kinase) causing glyceric aciduria were previously noted. Since glycine changes were not observed in almost all of the subsequently reported cases of d-glyceric aciduria, this theory of NKH as a secondary syndrome of d-glyceric aciduria was revisited in this work. We showed that this historic patient harbored a homozygous missense mutation in AMT c.350C>T, p.Ser117Leu, and enzymatic assay of the expressed mutation confirmed the pathogeneity of the p.Ser117Leu mutation. We conclude that the original d-glyceric aciduria patient also had classic NKH and that this co-occurrence of two inborn errors of metabolism explains the original presentation. We conclude that no evidence remains that d-glyceric aciduria would cause NKH.


Assuntos
Ácidos Glicéricos/urina , Hiperglicinemia não Cetótica/complicações , Hiperoxalúria Primária/complicações , Hiperoxalúria Primária/genética , Aminoácido Oxirredutases/genética , Aminoácido Oxirredutases/metabolismo , Aminometiltransferase/genética , Proteínas de Transporte/genética , Proteínas de Transporte/metabolismo , Diagnóstico Diferencial , Epilepsia , Ácidos Glicéricos/metabolismo , Glicina/metabolismo , Homozigoto , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Hiperglicinemia não Cetótica/etiologia , Hiperglicinemia não Cetótica/genética , Hiperoxalúria Primária/diagnóstico , Masculino , Complexos Multienzimáticos/genética , Complexos Multienzimáticos/metabolismo , Mutação de Sentido Incorreto , Fosfotransferases (Aceptor do Grupo Álcool)/deficiência , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Transferases/genética , Transferases/metabolismo
19.
Neurosciences (Riyadh) ; 22(2): 131-133, 2017 04.
Artigo em Inglês | MEDLINE | ID: mdl-28416785

RESUMO

Non-ketotic hyperglycinemia (NKH) is a rare inborn error of metabolism and is caused by a glycine cleavage system deficiency. Eighty-five percent of patients present with the neonatal type of NKH, the infants initially develop lethargy, seizures, and episodes of apnea, and most often death. Between 60-90% of cases are caused by mutations in the glycine decarboxylase (GLDC). We believed that more mutation reports especially for rare disease as NKH help to evaluate the genotype-phenotype relationship in patients with GLDC. In this study, we describe a case of a neonate admitted to intensive care unit with hypotonia, respiratory failure, lethargy, poor feeding. Due to the history of 2 non-ketotic hyperglycinemia diagnosed male siblings, molecular prenatal diagnosis in patient was performed and a novel c.2963G>A (Arg998Gln) homozygous mutation within the GLDC gene has been detected. We aimed to contribute to mutation knowledge pool of GLDC gene with a novel mutation.


Assuntos
Glicina Desidrogenase (Descarboxilante)/genética , Hiperglicinemia não Cetótica/genética , Mutação/genética , Saúde da Família , Feminino , Humanos , Lactente
20.
Zhongguo Dang Dai Er Ke Za Zhi ; 19(3): 268-271, 2017 Mar.
Artigo em Zh | MEDLINE | ID: mdl-28302194

RESUMO

Nonketotic hyperglycinemia (NKH) is a rare, inborn error of metabolism. In this case report, a Chinese male infant was diagnosed with NKH caused by GLDC gene mutation. The clinical characteristics and genetic diagnosis were reported. The infant presented with an onset of early metabolic encephalopathy and Ohtahara syndrome. Both blood and urinary levels of metabolites were in the normal range. Brain MRI images indicated a poor development of corpus callosum, and a burst suppression pattern was found in the EEG. Results of target gene sequencing technology combined with multiplex ligation-dependent probe amplification (MLPA) indicated a heterozygous missense mutation of c.1786 C>T (p.R596X) in maternal exon 15 and a loss of heterozygosity of 4-15 exon gross deletions in paternal GLDC gene. These definite pathogenic mutations confirmed the diagnosis of NKH. The infant's clinical condition was not improved after treatment with adreno-cortico-tropic-hormone, topiramate and dextromethorphan, and he finally died at 4 months of age. Patients with NKH often exhibit complicated clinical phenotypes and are lack of specific symptoms. NKH could be diagnosed by metabolic screening and molecular genetic analysis.


Assuntos
Hiperglicinemia não Cetótica/genética , Glicina Desidrogenase (Descarboxilante)/genética , Humanos , Hiperglicinemia não Cetótica/diagnóstico , Recém-Nascido , Masculino , Mutação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA