Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 313
Filter
1.
Mol Genet Metab ; 134(1-2): 156-163, 2021.
Article in English | MEDLINE | ID: mdl-34556413

ABSTRACT

Acyl CoA Dehydrogenase 9 (ACAD9) is a member of the family of flavoenzymes that catalyze the dehydrogenation of acyl-CoAs to 2,3 enoyl-CoAs in mitochondrial fatty acid oxidation (FAO). Inborn errors of metabolism of all family members, including ACAD9, have been described in humans, and represent significant causes of morbidity and mortality particularly in children. ACAD9 deficiency leads to a combined defect in fatty acid oxidation and oxidative phosphorylation (OXPHOS) due to a dual role in the pathways. In addition to its function in mitochondrial FAO, ACAD9 has a second function as one of 14 factors responsible for assembly of complex I of the electron transport chain (ETC). Considerable controversy remains over the relative role of these two functions in normal physiology and the disparate clinical findings described in patients with ACAD9 deficiency. To better understand the normal function of ACAD9 and the pathophysiology of its deficiency, several knock out mouse models were developed. Homozygous total body knock out appeared to be lethal as no ACAD9 animals were obtained. Cre-lox technology was then used to generate tissue-specific deletion of the gene. Cardiac-specific ACAD9 deficient animals had severe neonatal cardiomyopathy and died by 17 days of age. They had severe mitochondrial dysfunction in vitro. Muscle-specific mutants were viable but exhibited muscle weakness. Additional studies of heart muscle from the cardiac specific deficient animals were used to examine the evolutionarily conserved signaling Intermediate in toll pathway (ECSIT) protein, a known binding partner of ACAD9 in the electron chain complex I assembly pathway. As expected, ECSIT levels were significantly reduced in the absence of ACAD9 protein, consistent with the demonstrated impairment of the complex I assembly. The various ACAD9 deficient animals should serve as useful models for development of novel therapeutics for this disorder.


Subject(s)
Acidosis/genetics , Acidosis/physiopathology , Acyl-CoA Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/physiopathology , Cardiomyopathy, Hypertrophic/genetics , Cardiomyopathy, Hypertrophic/physiopathology , Disease Models, Animal , Mice , Mitochondrial Diseases/genetics , Mitochondrial Diseases/physiopathology , Muscle Weakness/genetics , Muscle Weakness/physiopathology , Acidosis/complications , Acyl-CoA Dehydrogenase/genetics , Adaptor Proteins, Signal Transducing/metabolism , Amino Acid Metabolism, Inborn Errors/complications , Animals , Cardiomyopathies/etiology , Cardiomyopathies/genetics , Cardiomyopathy, Hypertrophic/complications , Electron Transport Complex I/genetics , Mitochondrial Diseases/complications , Muscle Weakness/complications , Mutation
2.
J Inherit Metab Dis ; 44(1): 9-21, 2021 01.
Article in English | MEDLINE | ID: mdl-32412122

ABSTRACT

Organic acidurias (OADs) comprise a biochemically defined group of inherited metabolic diseases. Increasing awareness, reliable diagnostic work-up, newborn screening programs for some OADs, optimized neonatal and intensive care, and the development of evidence-based recommendations have improved neonatal survival and short-term outcome of affected individuals. However, chronic progression of organ dysfunction in an aging patient population cannot be reliably prevented with traditional therapeutic measures. Evidence is increasing that disease progression might be best explained by mitochondrial dysfunction. Previous studies have demonstrated that some toxic metabolites target mitochondrial proteins inducing synergistic bioenergetic impairment. Although these potentially reversible mechanisms help to understand the development of acute metabolic decompensations during catabolic state, they currently cannot completely explain disease progression with age. Recent studies identified unbalanced autophagy as a novel mechanism in the renal pathology of methylmalonic aciduria, resulting in impaired quality control of organelles, mitochondrial aging and, subsequently, progressive organ dysfunction. In addition, the discovery of post-translational short-chain lysine acylation of histones and mitochondrial enzymes helps to understand how intracellular key metabolites modulate gene expression and enzyme function. While acylation is considered an important mechanism for metabolic adaptation, the chronic accumulation of potential substrates of short-chain lysine acylation in inherited metabolic diseases might exert the opposite effect, in the long run. Recently, changed glutarylation patterns of mitochondrial proteins have been demonstrated in glutaric aciduria type 1. These new insights might bridge the gap between natural history and pathophysiology in OADs, and their exploitation for the development of targeted therapies seems promising.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/therapy , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Brain/metabolism , Brain/pathology , Brain Diseases, Metabolic, Inborn/metabolism , Brain Diseases, Metabolic, Inborn/pathology , Energy Metabolism , Humans , Infant, Newborn , Methylmalonic Acid/metabolism , Neonatal Screening
3.
J Cell Mol Med ; 24(1): 921-929, 2020 01.
Article in English | MEDLINE | ID: mdl-31705628

ABSTRACT

y+LAT1 (encoded by SLC7A7), together with y+LAT2 (encoded by SLC7A6), is the alternative light subunits composing the heterodimeric transport system y+L for cationic and neutral amino acids. SLC7A7 mutations cause lysinuric protein intolerance (LPI), an inherited multisystem disease characterized by low plasma levels of arginine and lysine, protein-rich food intolerance, failure to thrive, hepatosplenomegaly, osteoporosis, lung involvement, kidney failure, haematologic and immunological disorders. The reason for the heterogeneity of LPI symptoms is thus far only poorly understood. Here, we aimed to quantitatively compare the expression of SLC7A7 and SLC7A6 among different human cell types and evaluate y+LAT1 and y+LAT2 contribution to arginine transport. We demonstrate that system y+L-mediated arginine transport is mainly accounted for by y+LAT1 in monocyte-derived macrophages (MDM) and y+LAT2 in fibroblasts. The kinetic analysis of arginine transport indicates that y+LAT1 and y+LAT2 share a comparable affinity for the substrate. Differences have been highlighted in the expression of SLC7A6 and SLC7A7 mRNA among different cell models: while SLC7A6 is almost equally expressed, SLC7A7 is particularly abundant in MDM, intestinal Caco-2 cells and human renal proximal tubular epithelial cells (HRPTEpC). The characterization of arginine uptake demonstrates that system y+L is operative in renal cells and in Caco-2 where, at the basolateral side, it mediates arginine efflux in exchange with leucine plus sodium. These findings explain the defective absorption/reabsorption of arginine in LPI. Moreover, y+LAT1 is the prevailing transporter in MDM sustaining a pivotal role in the pathogenesis of immunological complications associated with the disease.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Transport System y+L/metabolism , Amino Acid Transport Systems, Basic/metabolism , Arginine/metabolism , Fibroblasts/metabolism , Kidney Tubules/metabolism , Macrophages/metabolism , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Transport System y+L/genetics , Amino Acid Transport Systems, Basic/genetics , Biological Transport , Caco-2 Cells , Humans , Lysine/metabolism , Mutation , Sodium/metabolism
4.
BMC Med Genet ; 21(1): 22, 2020 02 03.
Article in English | MEDLINE | ID: mdl-32013889

ABSTRACT

BACKGROUND: Methylmalonic acidemia (MMA), which is an autosomal recessive metabolic disorder, is caused by mutations in methylmalonyl-CoA mutase (MUT) gene. As a result, the conversion of methylmalonyl-CoA to succinyl-CoA is impaired in this disorder, leading to a wide range of clinical manifestations varying from no signs or symptoms to severe lethargy and metabolic crisis in newborn infants. Since identification of novel mutations in MUT gene can help discover the exact pathogenesis of MMA and also use these disease-causing mutations in prenatal diagnosis, this study was conducted to uncover the possible mutations in an Iranian couple with a deceased offspring clinically diagnosed as having organic acidemia. Moreover, to prevent the occurrence of the mutation in the next pregnancy, we took the advantage of pre-implantation genetic diagnosis (PGD), which resulted in a successful pregnancy. CASE PRESENTATION: The affected individual was a 15-month-old boy who passed away due to aspiration pneumonia. The child presented at the age of 3 months with lethargy, protracted vomiting, hypotonia, and decreased level of consciousness. To find the mutated gene, Next Generation Sequencing (NGS) was performed as carrier testing for the parents and the results revealed a novel (private) heterozygous missense mutation in MUT gene (c.1055A > G, p.Q352R). After performing PGD on three blastomeres, one was identified as being homozygous wild-type that was followed by successful pregnancy. CONCLUSIONS: Our study identified a novel, deleterious, heterozygous missense mutation in MUT gene in a couple and helps to consider the genetic counselling and prenatal diagnosis more seriously for this family with clinical phenotypes of organic acidemia.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Methylmalonyl-CoA Mutase/genetics , Preimplantation Diagnosis , Acyl Coenzyme A/genetics , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/physiopathology , Child , Female , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Infant , Infant, Newborn , Iran , Male , Mutation, Missense/genetics , Phenotype , Pregnancy
5.
J Hum Genet ; 65(9): 759-769, 2020 Sep.
Article in English | MEDLINE | ID: mdl-32409695

ABSTRACT

Aromatic L-amino acid decarboxylase deficiency (AADCD) is a rare neurotransmitter metabolic disorder caused by DDC gene mutations, which leads to the metabolic disturbance of dopamine and serotonin. Most of the reported cases came from Taiwan China, but patients from mainland China were seldomly reported. The current study was the largest AADCD patient cohort from mainland China. Twenty-three patients with clinical features of AADCD and DDC gene variants were recruited. A total of 16 DDC variants were identified in this study, of which four variants (c.2T>C, c.277A>G, c.1021+1G>A, c.565G>T) were never reported previously. The intronic variant c.714+4A>T was the most common one, with an allele frequency of 45.7%. And patients carried this intronic variant presented with severe clinical manifestations, all of whom were bedridden. In this study, the average onset age was 3.61 ± 1.28 months and the average age of diagnosis was 12.91 ± 5.62 months. Early onset hypotonia, oculogyric crises, and autonomic symptoms such as excessive sweating, nasal congestion and profuse nasal, and oropharyngeal secretions, were common in our patients. Eighteen patients (78.3%) got various degree of improvement after using pyridoxine monotherapy or different combination of pyridoxine, dopamine agonists, and monoamine oxidase (MAO) inhibitors.


Subject(s)
Amino Acid Metabolism, Inborn Errors/epidemiology , Amino Acid Metabolism, Inborn Errors/genetics , Aromatic-L-Amino-Acid Decarboxylases/deficiency , Genetic Predisposition to Disease/genetics , Alleles , Amino Acid Metabolism, Inborn Errors/drug therapy , Amino Acid Metabolism, Inborn Errors/physiopathology , Aromatic-L-Amino-Acid Decarboxylases/chemistry , Aromatic-L-Amino-Acid Decarboxylases/genetics , China , Cohort Studies , Demography , Dopamine Agonists/therapeutic use , Exons , Female , Genetic Variation , Heterozygote , Homozygote , Humans , Infant , Introns , Male , Monoamine Oxidase Inhibitors/therapeutic use , Muscle Hypotonia/complications , Muscle Hypotonia/genetics , Mutation , Pyridoxine/therapeutic use , Exome Sequencing
6.
Arch Biochem Biophys ; 696: 108646, 2020 12 15.
Article in English | MEDLINE | ID: mdl-33098870

ABSTRACT

Organic acidurias (OADs) are inherited disorders of amino acid metabolism biochemically characterized by accumulation of short-chain carboxylic acids in tissues and biological fluids of the affected patients and clinically by predominant neurological manifestations. Some of these disorders are amenable to treatment, which significantly decreases mortality and morbidity, but it is still ineffective to prevent long-term neurologic and systemic complications. Although pathogenesis of OADs is still poorly established, recent human and animal data, such as lactic acidosis, mitochondrial morphological alterations, decreased activities of respiratory chain complexes and altered parameters of oxidative stress, found in tissues from patients and from genetic mice models with these diseases indicate that disruption of critical mitochondrial functions and oxidative stress play an important role in their pathophysiology. Furthermore, organic acids that accumulate in the most prevalent OADs were shown to compromise bioenergetics, by decreasing ATP synthesis, mitochondrial membrane potential, reducing equivalent content and calcium retention capacity, besides inducing mitochondrial swelling, reactive oxygen and nitrogen species generation and apoptosis. It is therefore presumed that secondary mitochondrial dysfunction and oxidative stress caused by major metabolites accumulating in OADs contribute to tissue damage in these pathologies.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/physiopathology , Mitochondria/metabolism , Nervous System Diseases/etiology , Nervous System Diseases/physiopathology , Oxidative Stress/physiology , Animals , Brain/metabolism , Carboxylic Acids/metabolism , Humans
7.
J Inherit Metab Dis ; 43(2): 200-215, 2020 03.
Article in English | MEDLINE | ID: mdl-31603991

ABSTRACT

Glutamate is involved in a variety of metabolic pathways. We reviewed the literature on genetic defects of enzymes that directly metabolise glutamate, leading to inborn errors of glutamate metabolism. Seventeen genetic defects of glutamate metabolising enzymes have been reported, of which three were only recently identified. These 17 defects affect the inter-conversion of glutamine and glutamate, amino acid metabolism, ammonia detoxification, and glutathione metabolism. We provide an overview of the clinical and biochemical phenotypes of these rare defects in an effort to ease their recognition. By categorising these by biochemical pathway, we aim to create insight into the contributing role of deviant glutamate and glutamine levels to the pathophysiology. For those disorders involving the inter-conversion of glutamine and glutamate, these deviant levels are postulated to play a pivotal pathophysiologic role. For the other IEM however-with the exception of urea cycle defects-abnormal glutamate and glutamine concentrations were rarely reported. To create insight into the clinical consequences of disturbed glutamate metabolism-rather than individual glutamate and glutamine levels-the prevalence of phenotypic abnormalities within the 17 IEM was compared to their prevalence within all Mendelian disorders and subsequently all disorders with metabolic abnormalities notated in the Human Phenotype Ontology (HPO) database. For this, a hierarchical database of all phenotypic abnormalities of the 17 defects in glutamate metabolism based on HPO was created. A neurologic phenotypic spectrum of developmental delay, ataxia, seizures, and hypotonia are common in the inborn errors of enzymes in glutamate metabolism. Additionally, ophthalmologic and skin abnormalities are often present, suggesting that disturbed glutamate homeostasis affects tissues of ectodermal origin: brain, eye, and skin. Reporting glutamate and glutamine concentrations in patients with inborn errors of glutamate metabolism would provide additional insight into the pathophysiology.


Subject(s)
Amino Acid Metabolism, Inborn Errors/enzymology , Glutamates/metabolism , Glutamine/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Databases, Factual , Deficiency Diseases/etiology , Glutamates/deficiency , Glutamine/deficiency , Humans
8.
J Inherit Metab Dis ; 43(5): 1121-1130, 2020 09.
Article in English | MEDLINE | ID: mdl-32369189

ABSTRACT

Aromatic l-amino acid decarboxylase deficiency (AADCD) is a rare, autosomal recessive neurodevelopmental disorder characterized by impaired synthesis of dopamine, noradrenaline, adrenaline and serotonin, leading to a complex syndrome of motor, behavioral, and autonomic symptoms. This retrospective study assessed the symptoms and developmental outcome of a large international cohort of patients with AADCD via physician and/or caregiver responses to a detailed, standardized questionnaire. Sixty-three patients (60% female; ages 6 months-36 years, median 7 years; 58 living) from 23 individual countries participated. Common symptoms at onset (median age 3 months, range 0-12 months) were hypotonia, developmental delay, and/or oculogyric crises. Oculogyric crises were present in 97% of patients aged 2 to 12 years, occurred in the majority of patients in all age groups, and tended to be most severe during early childhood. Prominent non-motor symptoms were sleep disturbance, irritable mood, and feeding difficulties. The majority of subjects (70%) had profound motor impairment characterized by absent head control and minimal voluntary movement, while 17% had mild motor impairment and were able to walk independently. Dopamine agonists were the medications most likely to produce some symptomatic benefit, but were associated with dose-limiting side effects (dyskinesia, insomnia, irritability, vomiting) that led to discontinuation 25% of the time. The age distribution of our cohort (70% of subjects under age 13 years) and the observation of a greater proportion of patients with a more severe disease phenotype in the younger compared to the older patients, both suggest a significant mortality risk during childhood for patients with severe disease.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/therapy , Aromatic-L-Amino-Acid Decarboxylases/deficiency , Dopamine Agonists/therapeutic use , Dystonic Disorders/etiology , Ocular Motility Disorders/etiology , Adolescent , Adult , Age Factors , Child , Child, Preschool , Dopamine Agonists/adverse effects , Female , Genetic Therapy , Humans , Infant , Internationality , Male , Phenotype , Retrospective Studies , Young Adult
9.
J Inherit Metab Dis ; 43(3): 424-437, 2020 05.
Article in English | MEDLINE | ID: mdl-31828787

ABSTRACT

Evidence for effectiveness of newborn screening (NBS) for propionic acidemia (PA) and isolated methylmalonic acidemia (MMA) is scarce. Prior to implementation in the Netherlands, we aim to estimate the expected health gain of NBS for PA and MMA. In this national retrospective cohort study, the clinical course of 76/83 Dutch PA and MMA patients, diagnosed between January 1979 and July 2019, was evaluated. Five clinical outcome parameters were defined: adverse outcome of the first symptomatic phase, frequency of acute metabolic decompensations (AMD), cognitive function, mitochondrial complications, and treatment-related complications. Outcomes of patients identified by family testing were compared with the outcomes of their index siblings. An adverse outcome due to the first symptomatic phase was recorded in 46% of the clinically diagnosed patients. Outcome of the first symptomatic phase was similar in 5/9 sibling pairs and better in 4/9 pairs. Based on the day of diagnosis of the clinically diagnosed patients and sibling pair analysis, a preliminary estimated reduction of adverse outcome due to the first symptomatic phase from 46% to 36%-38% was calculated. Among the sibling pairs, AMD frequency, cognitive function, mitochondrial, and treatment-related complications were comparable. These results suggest that the health gain of NBS for PA and MMA in overall outcome may be limited, as only a modest decrease of adverse outcomes due to the first symptomatic phase is expected. With current clinical practice, no reduced AMD frequency, improved cognitive function, or reduced frequency of mitochondrial or treatment-related complications can be expected.


Subject(s)
Amino Acid Metabolism, Inborn Errors/diagnosis , Mitochondrial Diseases/complications , Propionic Acidemia/diagnosis , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/therapy , Cognition , Female , Humans , Infant, Newborn , Kaplan-Meier Estimate , Male , Methylmalonic Acid , Mitochondrial Diseases/physiopathology , Neonatal Screening , Netherlands , Propionic Acidemia/physiopathology , Propionic Acidemia/therapy , Retrospective Studies , Siblings
10.
Neuropediatrics ; 51(1): 6-21, 2020 02.
Article in English | MEDLINE | ID: mdl-31634934

ABSTRACT

Inherited neurotransmitter disorders are rare neurometabolic conditions which encompass genetic disorders of neurotransmitter metabolism or transport. The clinical manifestations of these rare disorders are often nonspecific, ranging from encephalopathies and seizures to movement disorders. As a consequence, neurotransmitter disorders are underrecognized and often misdiagnosed. Accurate and timely diagnosis is, however, of utmost importance, given the availability of therapeutic strategies. A high index of clinical suspicion and familiarity with the neuroimaging phenotypes is therefore crucial. While the imaging features of various neurotransmitter disorders often overlap and are nonspecific, imaging can be helpful in providing useful clues to guide the diagnostic algorithm for uncommon conditions in a neonate presenting with nonspecific neurological symptoms. In this review paper, we aim to bring together current knowledge of neuroimaging phenotypes associated with inherited (primary) disorders of neurotransmitter biosynthesis. Magnetic resonance imaging phenotypes of disorders of monoamine biosynthesis, primary cerebral folate deficiency, disorders of pyridoxine metabolism, disorders of gamma-aminobutyric acid metabolism, nonketotic hyperglycinemia (glycine encephalopathy), disorders of serine biosynthesis, and cerebral creatine deficiency syndrome will be discussed and illustrated with case examples.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Biogenic Monoamines , Brain Diseases, Metabolic , Neuroimaging , Amino Acid Metabolism, Inborn Errors/diagnostic imaging , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Metabolism, Inborn Errors/physiopathology , Biogenic Monoamines/metabolism , Brain Diseases, Metabolic/diagnostic imaging , Brain Diseases, Metabolic/metabolism , Brain Diseases, Metabolic/pathology , Brain Diseases, Metabolic/physiopathology , Humans
11.
Mediators Inflamm ; 2020: 5785378, 2020.
Article in English | MEDLINE | ID: mdl-33144848

ABSTRACT

PURPOSE: Systemic mastocytosis (SM) is characterized by a clonal proliferation of neoplastic mast cells (MCs) in one or more extracutaneous organs including the bone marrow (BM). SM is often associated with osteoporosis (OP) and fractures. Hypertryptasemia usually occurs in SM. We investigated the prevalence of hypertryptasemia in a series of severe osteoporotic patients, the performance of the tryptase test in diagnosing SM in these patients, and their bone features. METHODS: The medical records of 232 patients (168 females and 64 males) with a diagnosis of OP (50.4% with fractures) and a serum tryptase assessment were reviewed. BM assessment was performed in a subset of hypertryptasemic patients; clinical, biochemical, and radiographic data were collected. RESULTS: Hypertryptasemia was detected in 33 patients. BM assessment (n = 16) was normal in 8 hypertryptasemic patients, while BM criteria for the diagnosis of SM were met in 3 patients, MC alterations were detected in 4 patients, and one patient presented a polycythemia vera. Serum tryptase levels were higher than 11.4 ng/ml in all patients with BM alterations. The best cut-off of tryptase level related to BM alterations was 17.9 ng/ml, with a sensibility and sensitivity of 75% (AUC = 0.797 and P = 0.015 by ROC analysis). All osteoporotic patients with hypertryptasemia experienced at least one vertebral fracture associated with a severe reduction of the lumbar bone mineral density. CONCLUSIONS: The prevalence of MC-related disorders in severe OP was 3.0%, accounting for the 7.4% of the secondary causes of OP. MC-related disorders may be involved in bone fragility and assessment of serum tryptase is useful to detect MC-related disorders.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Hypercalciuria/blood , Hypercalciuria/physiopathology , Mast Cells/pathology , Mastocytosis, Systemic/pathology , Adult , Aged , Amino Acid Metabolism, Inborn Errors/blood , Bone Marrow/metabolism , Bone Marrow/pathology , Female , Humans , Male , Mastocytosis, Systemic/blood , Middle Aged , Osteoporosis/blood , Osteoporosis/physiopathology , Tryptases/metabolism
12.
J Hum Genet ; 64(4): 305-312, 2019 Apr.
Article in English | MEDLINE | ID: mdl-30651581

ABSTRACT

Cobalamin G (cblG) and cobalamin J (cblJ) defects are rare disorders of cbl metabolism caused by MTR and ABCD4 mutations, respectively. Patients with atypical biochemical features can be missed by current newborn screening using tandem mass spectrometry (MS/MS), in which total homocysteine (tHCY) in dried blood spots (DBS) is not a primary biomarker. Two Chinese patients suspected of cbl defect but missed by newborn screening were studied. Using comprehensive metabolic analyses including MS/MS assay for tHCY in DBS, slightly low methionine in Patient 1, methymalonic aciduria in Patient 2, and homocysteinemia in both patients were detected, and DBS tHCY of two patients were obviously elevated (59.22 µmol/L, 17.75 µmol/L) compared to 140 healthy controls (2.5th-97.5th percentile, 1.05-8.22 µmol/L). Utilizing whole-exome sequencing, we found two novel MTR variants c.871C>T (p.Pro291Ser) and c.1771C>T (p.Arg591*) in Patient 1, and a ABCD4 homozygous variant c.423C>G (p.Asn141Lys) in Patient 2. Our study identified the first cblG patient and cblJ patient in mainland China, and highlighted comprehensive metabolic analyses and genetic tests in patients suspected of cbl defects. It also indicated that supplementary MS/MS assay for tHCY in DBS may be practical for early diagnosis of homocysteinemia, without repeated blood sampling.


Subject(s)
5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/genetics , ATP-Binding Cassette Transporters/genetics , Amino Acid Metabolism, Inborn Errors/blood , Neonatal Screening , Vitamin B 12/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/physiopathology , Child , Child, Preschool , China , Female , Homocysteine/blood , Humans , Infant , Infant, Newborn , Male , Mutation , Tandem Mass Spectrometry , Vitamin B 12/blood , Exome Sequencing
13.
Mov Disord ; 34(5): 598-613, 2019 05.
Article in English | MEDLINE | ID: mdl-30557456

ABSTRACT

There are several hundred single-gene disorders that we classify as inborn errors of metabolism. Inborn errors of metabolism are often rare and highly heterogeneous multisystem diseases with non-neurological and neurological manifestations, commonly with onset during childhood. Movement disorders are among the most common neurological problems in inborn errors of metabolism, but, in many cases, remain poorly defined. Although movement disorders are usually not the only and often not the presenting symptom, their recognition can facilitate a diagnosis. Movement disorders contribute substantially to the morbidity in inborn errors of metabolism and can have a significant impact on quality of life. Common metabolic movement disorders include the monoamine neurotransmitter disorders, disorders of amino and organic acid metabolism, metal storage disorders, lysosomal storage disorders, congenital disorders of autophagy, disorders of creatine metabolism, vitamin-responsive disorders, and disorders of energy metabolism. Importantly, disease-modifying therapies exist for a number of inborn errors of metabolism, and early recognition and treatment can prevent irreversible CNS damage and reduce morbidity and mortality. A phenomenology-based approach, based on the predominant movement disorder, can facilitate a differential diagnosis and can guide biochemical, molecular, and imaging testing. The complexity of metabolic movement disorders demands an interdisciplinary approach and close collaboration of pediatric neurologists, neurologists, geneticists, and experts in metabolism. In this review, we develop a general framework for a phenomenology-based approach to movement disorders in inborn errors of metabolism and discuss an approach to identifying the "top ten" of treatable inborn errors of metabolism that present with movement disorders-diagnoses that should never be missed. © 2018 International Parkinson and Movement Disorder Society.


Subject(s)
Metabolism, Inborn Errors/physiopathology , Movement Disorders/physiopathology , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/physiopathology , Amino Acid Metabolism, Inborn Errors/therapy , Ataxia/complications , Ataxia/diagnosis , Ataxia/etiology , Ataxia/physiopathology , Ataxia/therapy , Basal Ganglia Diseases/complications , Basal Ganglia Diseases/diagnosis , Basal Ganglia Diseases/physiopathology , Basal Ganglia Diseases/therapy , Brain Diseases, Metabolic/complications , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/physiopathology , Brain Diseases, Metabolic/therapy , Carbohydrate Metabolism, Inborn Errors/complications , Carbohydrate Metabolism, Inborn Errors/diagnosis , Carbohydrate Metabolism, Inborn Errors/physiopathology , Carbohydrate Metabolism, Inborn Errors/therapy , Chorea/etiology , Chorea/physiopathology , Dystonia/etiology , Dystonia/physiopathology , Dystonic Disorders/complications , Dystonic Disorders/diagnosis , Dystonic Disorders/etiology , Dystonic Disorders/physiopathology , Dystonic Disorders/therapy , Folic Acid Deficiency/complications , Folic Acid Deficiency/diagnosis , Folic Acid Deficiency/physiopathology , Folic Acid Deficiency/therapy , Glutaryl-CoA Dehydrogenase/deficiency , Hepatolenticular Degeneration/complications , Hepatolenticular Degeneration/diagnosis , Hepatolenticular Degeneration/physiopathology , Hepatolenticular Degeneration/therapy , Humans , Metabolic Diseases/complications , Metabolic Diseases/diagnosis , Metabolic Diseases/physiopathology , Metabolic Diseases/therapy , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/diagnosis , Metabolism, Inborn Errors/therapy , Monosaccharide Transport Proteins/deficiency , Movement Disorders/etiology , Muscle Spasticity/etiology
14.
Pediatr Res ; 86(1): 39-46, 2019 07.
Article in English | MEDLINE | ID: mdl-30986815

ABSTRACT

BACKGROUND: Environmental tobacco smoke (ETS) is a known risk factor for severe respiratory syncytial virus (RSV) infections, yet the mechanisms of ETS/RSV comorbidity are largely unknown. Cystathionine γ-lyase regulates important physiological functions of the respiratory tract. METHODS: We used mice genetically deficient in the cystathionine γ-lyase enzyme (CSE), the major H2S-generating enzyme in the lung to determine the contribution of H2S to airway disease in response to side-stream tobacco smoke (TS), and to TS/RSV co-exposure. RESULTS: Following a 2-week period of exposure to TS, CSE-deficient mice (KO) showed a dramatic increase in airway hyperresponsiveness (AHR) to methacholine challenge, and greater airway cellular inflammation, compared with wild-type (WT) mice. TS-exposed CSE KO mice that were subsequently infected with RSV exhibited a more severe clinical disease, airway obstruction and AHR, enhanced viral replication, and lung inflammation, compared with TS-exposed RSV-infected WT mice. TS-exposed RSV-infected CSE KO mice had also a significant increase in the number of neutrophils in bronchoalveolar lavage fluid and increased levels of inflammatory cytokines and chemokines. CONCLUSION: This study demonstrates the critical contribution of the H2S-generating pathway to airway reactivity and disease following exposure to ETS alone or in combination with RSV infection.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Cystathionine gamma-Lyase/deficiency , Lung/physiopathology , Lung/virology , Respiratory Hypersensitivity/complications , Respiratory Syncytial Virus Infections/complications , Tobacco Smoke Pollution/adverse effects , Animals , Bronchoalveolar Lavage Fluid , Cytokines/metabolism , Female , Genetic Predisposition to Disease , Hydrogen Sulfide/chemistry , Inflammation/etiology , Male , Methacholine Chloride , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophils/cytology , Respiratory Hypersensitivity/virology , Respiratory Syncytial Viruses
15.
J Inherit Metab Dis ; 42(5): 745-761, 2019 09.
Article in English | MEDLINE | ID: mdl-31119742

ABSTRACT

Despite realizing increased survival rates for propionic acidemia (PA) and methylmalonic acidemia (MMA) patients, the current therapeutic regimen is inadequate for preventing or treating the devastating complications that still can occur. The elucidation of pathophysiology of these complications allows us to evaluate and rethink treatment strategies. In this review we display and discuss potential therapy targets and we give a systematic overview on current, experimental and unexplored treatment strategies in order to provide insight in what we have to offer PA and MMA patients, now and in the future. Evidence on the effectiveness of treatment strategies is often scarce, since none were tested in randomized clinical trials. This raises concerns, since even the current consensus on best practice treatment for PA and MMA is not without controversy. To attain substantial improvements in overall outcome, gene, mRNA or enzyme replacement therapy is most promising since permanent reduction of toxic metabolites allows for a less strict therapeutic regime. Hereby, both mitochondrial-associated and therapy induced complications can theoretically be prevented. However, the road from bench to bedside is long, as it is challenging to design a drug that is delivered to the mitochondria of all tissues that require enzymatic activity, including the brain, without inducing any off-target effects. To improve survival rate and quality of life of PA and MMA patients, there is a need for systematic (re-)evaluation of accepted and potential treatment strategies, so that we can better determine who will benefit when and how from which treatment strategy.


Subject(s)
Amino Acid Metabolism, Inborn Errors/therapy , Mitochondrial Diseases/therapy , Propionic Acidemia/therapy , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Brain/metabolism , Disease Management , Humans , Mitochondria/metabolism , Mitochondrial Diseases/physiopathology , Propionic Acidemia/physiopathology
16.
J Inherit Metab Dis ; 42(5): 730-744, 2019 09.
Article in English | MEDLINE | ID: mdl-31119747

ABSTRACT

Over the last decades, advances in clinical care for patients suffering from propionic acidemia (PA) and isolated methylmalonic acidemia (MMA) have resulted in improved survival. These advances were possible thanks to new pathophysiological insights. However, patients may still suffer from devastating complications which largely determine the unsatisfying overall outcome. To optimize our treatment strategies, better insight in the pathophysiology of complications is needed. Here, we perform a systematic data-analysis of cohort studies and case-reports on PA and MMA. For each of the prevalent and rare complications, we summarize the current hypotheses and evidence for the underlying pathophysiology of that complication. A common hypothesis on pathophysiology of many of these complications is that mitochondrial impairment plays a major role. Assuming that complications in which mitochondrial impairment may play a role are overrepresented in monogenic mitochondrial diseases and, conversely, that complications in which mitochondrial impairment does not play a role are underrepresented in mitochondrial disease, we studied the occurrence of the complications in PA and MMA in mitochondrial and other monogenic diseases, using data provided by the Human Phenotype Ontology. Lastly, we combined this with evidence from literature to draw conclusions on the possible role of mitochondrial impairment in each complication. Altogether, this review provides a comprehensive overview on what we, to date, do and do not understand about pathophysiology of complications occurring in PA and MMA and about the role of mitochondrial impairment herein.


Subject(s)
Amino Acid Metabolism, Inborn Errors/complications , Mitochondria/pathology , Mitochondrial Diseases/etiology , Propionic Acidemia/complications , Amino Acid Metabolism, Inborn Errors/physiopathology , Animals , Humans , Mitochondrial Diseases/physiopathology , Propionic Acidemia/physiopathology
18.
Mol Genet Metab ; 124(3): 204-209, 2018 07.
Article in English | MEDLINE | ID: mdl-29789193

ABSTRACT

Patients with primary serine biosynthetic defects manifest with intellectual disability, microcephaly, ichthyosis, seizures and peripheral neuropathy. The underlying pathogenesis of peripheral neuropathy in these patients has not been elucidated, but could be related to a decrease in the availability of certain classical sphingolipids, or to an increase in atypical sphingolipids. Here, we show that patients with primary serine deficiency have a statistically significant elevation in specific atypical sphingolipids, namely deoxydihydroceramides of 18-22 carbons in acyl length. We also show that patients with aberrant plasma serine and alanine levels secondary to mitochondrial disorders also display peripheral neuropathy along with similar elevations of atypical sphingolipids. We hypothesize that the etiology of peripheral neuropathy in patients with primary mitochondrial disorders is related to this elevation of deoxysphingolipids, in turn caused by increased availability of alanine and decreased availability of serine. These findings could have important therapeutic implications for the management of these patients.


Subject(s)
Amino Acid Metabolism, Inborn Errors/physiopathology , Mitochondrial Diseases/physiopathology , Serine/deficiency , Sphingolipids/metabolism , Adolescent , Adult , Aged , Case-Control Studies , Child , Female , Humans , Male , Middle Aged , Prognosis , Young Adult
19.
Mol Genet Metab ; 124(4): 266-277, 2018 08.
Article in English | MEDLINE | ID: mdl-29934063

ABSTRACT

BACKGROUND: Methylmalonic aciduria (MMAuria) is an inborn error of metabolism leading to neurological deterioration. In this study, we used 3D organotypic brain cell cultures derived from embryos of a brain-specific Mut-/- (brain KO) mouse to investigate mechanisms leading to brain damage. We challenged our in vitro model by a catabolic stress (temperature shift). RESULTS: Typical metabolites for MMAuria as well as a massive NH4+ increase were found in the media of brain KO cultures. We investigated different pathways of intracerebral NH4+ production and found increased expression of glutaminase 2 and diminished expression of GDH1 in Mut-/- aggregates. While all brain cell types appeared affected in their morphological development in Mut-/- aggregates, the most pronounced effects were observed on astrocytes showing swollen fibers and cell bodies. Inhibited axonal elongation and delayed myelination of oligodendrocytes were also noted. Most effects were even more pronounced after 48 h at 39 °C. Microglia activation and an increased apoptosis rate suggested degeneration of Mut-/- brain cells. NH4+ accumulation might be the trigger for all observed alterations. We also found a generalized increase of chemokine concentrations in Mut-/- culture media at an early developmental stage followed by a decrease at a later stage. CONCLUSION: We proved for the first time that Mut-/- brain cells are indeed able to produce the characteristic metabolites of MMAuria. We confirmed significant NH4+ accumulation in culture media of Mut-/- aggregates, suggesting that intracellular NH4+ concentrations might even be higher, gave first clues on the mechanisms leading to NH4+ accumulation in Mut-/- brain cells, and showed the involvement of neuroinflammatory processes in the neuropathophysiology of MMAuria.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Ammonium Compounds/metabolism , Brain/metabolism , Methylmalonyl-CoA Mutase/genetics , Amino Acid Metabolism, Inborn Errors/metabolism , Amino Acid Metabolism, Inborn Errors/physiopathology , Ammonium Compounds/toxicity , Animals , Brain/physiopathology , Brain Injuries/genetics , Brain Injuries/metabolism , Brain Injuries/physiopathology , Humans , Methylmalonic Acid/metabolism , Mice , Mice, Knockout , Organ Culture Techniques
20.
Mol Genet Metab ; 125(3): 276-280, 2018 11.
Article in English | MEDLINE | ID: mdl-30217722

ABSTRACT

Three young patients with glutaric aciduria type I (age 6-23 years) of different ethnic origins, treated for their metabolic disease since early childhood, presented with malignant central nervous system tumors. We recommend continuing clinical follow-up, including monitoring of neurological manifestations and neuroradiological findings, in all patients with glutaric aciduria type I beyond early childhood, especially if adherence to diet is poor or the treatment was not started neonatally.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Brain Neoplasms/genetics , Brain/metabolism , Glioblastoma/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Adult , Amino Acid Metabolism, Inborn Errors/complications , Amino Acid Metabolism, Inborn Errors/diagnostic imaging , Amino Acid Metabolism, Inborn Errors/physiopathology , Brain/diagnostic imaging , Brain/pathology , Brain Diseases, Metabolic/complications , Brain Diseases, Metabolic/diagnostic imaging , Brain Diseases, Metabolic/physiopathology , Brain Neoplasms/complications , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/physiopathology , Child , Child, Preschool , Female , Glioblastoma/complications , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/genetics , Humans , Male , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL