Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 46
1.
J Community Genet ; 14(6): 613-620, 2023 Dec.
Article En | MEDLINE | ID: mdl-37847346

Newborn screening in Alaska includes screening for carnitine palmitoyltransferase 1A (CPT1A) deficiency. The CPT1A Arctic variant is a variant highly prevalent among Indigenous peoples in the Arctic. In this study, we sought to elicit Alaska Native (AN) community member and AN-serving healthcare providers' knowledge and perspectives on the CPT1A Arctic variant. Focus groups with community members and healthcare providers were held in two regions of Alaska between October 2018 and January 2019. Thematic analysis was used to identify recurring constructs. Knowledge and understanding about the CPT1A Arctic variant and its health impact varied, and participants were interested in learning more about it. Additional education for healthcare professionals was recommended to improve providers' ability to communicate with family caregivers about the Arctic variant. Engagement with AN community members identified opportunities to improve educational outreach via multiple modalities for providers and caregivers on the Arctic variant, which could help to increase culturally relevant guidance and avoid stigmatization, undue worry, and unnecessary intervention. Education and guidance on the care of infants and children homozygous for the CPT1A Arctic variant could improve care and reduce negative psychosocial effects.

2.
Sci Data ; 8(1): 114, 2021 04 21.
Article En | MEDLINE | ID: mdl-33883556

Every year individuals experience symptoms that remain undiagnosed by healthcare providers. In the United States, these rare diseases are defined as a condition that affects fewer than 200,000 individuals. However, there are an estimated 7000 rare diseases, and there are an estimated 25-30 million Americans in total (7.6-9.2% of the population as of 2018) affected by such disorders. The NIH Common Fund Undiagnosed Diseases Network (UDN) seeks to provide diagnoses for individuals with undiagnosed disease. Mass spectrometry-based metabolomics and lipidomics analyses could advance the collective understanding of individual symptoms and advance diagnoses for individuals with heretofore undiagnosed disease. Here, we report the mass spectrometry-based metabolomics and lipidomics analyses of blood plasma, urine, and cerebrospinal fluid from 148 patients within the UDN and their families, as well as from a reference population of over 100 individuals with no known metabolic diseases. The raw and processed data are available to the research community so that they might be useful in the diagnoses of current or future patients suffering from undiagnosed disorders.


Lipidomics , Metabolic Diseases/diagnosis , Metabolomics , Undiagnosed Diseases/diagnosis , Adolescent , Adult , Child , Child, Preschool , Datasets as Topic , Female , Humans , Infant , Infant, Newborn , Male , Mass Spectrometry , Metabolic Diseases/blood , Metabolic Diseases/cerebrospinal fluid , Metabolic Diseases/urine , Middle Aged , Undiagnosed Diseases/blood , Undiagnosed Diseases/cerebrospinal fluid , Undiagnosed Diseases/urine , Young Adult
4.
PLoS Genet ; 16(6): e1008841, 2020 06.
Article En | MEDLINE | ID: mdl-32544203

Hypomyelination, a neurological condition characterized by decreased production of myelin sheets by glial cells, often has no known etiology. Elucidating the genetic causes of hypomyelination provides a better understanding of myelination, as well as means to diagnose, council, and treat patients. Here, we present evidence that YIPPEE LIKE 3 (YPEL3), a gene whose developmental role was previously unknown, is required for central and peripheral glial cell development. We identified a child with a constellation of clinical features including cerebral hypomyelination, abnormal peripheral nerve conduction, hypotonia, areflexia, and hypertrophic peripheral nerves. Exome and genome sequencing revealed a de novo mutation that creates a frameshift in the open reading frame of YPEL3, leading to an early stop codon. We used zebrafish as a model system to validate that YPEL3 mutations are causative of neuropathy. We found that ypel3 is expressed in the zebrafish central and peripheral nervous system. Using CRISPR/Cas9 technology, we created zebrafish mutants carrying a genomic lesion similar to that of the patient. Our analysis revealed that Ypel3 is required for development of oligodendrocyte precursor cells, timely exit of the perineurial glial precursors from the central nervous system (CNS), formation of the perineurium, and Schwann cell maturation. Consistent with these observations, zebrafish ypel3 mutants have metabolomic signatures characteristic of oligodendrocyte and Schwann cell differentiation defects, show decreased levels of Myelin basic protein in the central and peripheral nervous system, and develop defasciculated peripheral nerves. Locomotion defects were observed in adult zebrafish ypel3 mutants. These studies demonstrate that Ypel3 is a novel gene required for perineurial cell development and glial myelination.


Gene Expression Regulation, Developmental , Hereditary Central Nervous System Demyelinating Diseases/genetics , Myelin Sheath/pathology , Neurogenesis/genetics , Tumor Suppressor Proteins/genetics , Animals , Brachial Plexus/diagnostic imaging , Child , DNA Mutational Analysis , Disease Models, Animal , Embryo, Nonmammalian , Female , Frameshift Mutation , Gray Matter/diagnostic imaging , Hereditary Central Nervous System Demyelinating Diseases/diagnostic imaging , Hereditary Central Nervous System Demyelinating Diseases/pathology , Humans , Magnetic Resonance Imaging , Neuroglia/pathology , Oligodendroglia , Sciatic Nerve/diagnostic imaging , White Matter/diagnostic imaging , Exome Sequencing , Zebrafish , Zebrafish Proteins/genetics
5.
Anal Chem ; 92(2): 1796-1803, 2020 01 21.
Article En | MEDLINE | ID: mdl-31742994

Advancements in molecular separations coupled with mass spectrometry have enabled metabolome analyses for clinical cohorts. A population of interest for metabolome profiling is patients with rare disease for which abnormal metabolic signatures may yield clues into the genetic basis, as well as mechanistic drivers of the disease and possible treatment options. We undertook the metabolome profiling of a large cohort of patients with mysterious conditions characterized through the Undiagnosed Diseases Network (UDN). Due to the size and enrollment procedures, collection of the metabolomes for UDN patients took place over 2 years. We describe the study designed to adjust for measurements collected over a long time scale and how this enabled statistical analyses to summarize the metabolome of individual patients. We demonstrate the removal of time-based batch effects, overall statistical characteristics of the UDN population, and two case studies of interest that demonstrate the utility of metabolome profiling for rare diseases.


Lipids/analysis , Models, Statistical , Undiagnosed Diseases/diagnosis , Cohort Studies , Humans , Metabolomics , Undiagnosed Diseases/metabolism
6.
N Engl J Med ; 379(22): 2131-2139, 2018 11 29.
Article En | MEDLINE | ID: mdl-30304647

BACKGROUND: Many patients remain without a diagnosis despite extensive medical evaluation. The Undiagnosed Diseases Network (UDN) was established to apply a multidisciplinary model in the evaluation of the most challenging cases and to identify the biologic characteristics of newly discovered diseases. The UDN, which is funded by the National Institutes of Health, was formed in 2014 as a network of seven clinical sites, two sequencing cores, and a coordinating center. Later, a central biorepository, a metabolomics core, and a model organisms screening center were added. METHODS: We evaluated patients who were referred to the UDN over a period of 20 months. The patients were required to have an undiagnosed condition despite thorough evaluation by a health care provider. We determined the rate of diagnosis among patients who subsequently had a complete evaluation, and we observed the effect of diagnosis on medical care. RESULTS: A total of 1519 patients (53% female) were referred to the UDN, of whom 601 (40%) were accepted for evaluation. Of the accepted patients, 192 (32%) had previously undergone exome sequencing. Symptoms were neurologic in 40% of the applicants, musculoskeletal in 10%, immunologic in 7%, gastrointestinal in 7%, and rheumatologic in 6%. Of the 382 patients who had a complete evaluation, 132 received a diagnosis, yielding a rate of diagnosis of 35%. A total of 15 diagnoses (11%) were made by clinical review alone, and 98 (74%) were made by exome or genome sequencing. Of the diagnoses, 21% led to recommendations regarding changes in therapy, 37% led to changes in diagnostic testing, and 36% led to variant-specific genetic counseling. We defined 31 new syndromes. CONCLUSIONS: The UDN established a diagnosis in 132 of the 382 patients who had a complete evaluation, yielding a rate of diagnosis of 35%. (Funded by the National Institutes of Health Common Fund.).


Genetic Testing , Rare Diseases/genetics , Sequence Analysis, DNA , Adult , Animals , Child , Diagnosis, Differential , Drosophila , Exome , Female , Genetic Testing/economics , Health Care Costs/statistics & numerical data , Humans , Male , Models, Animal , National Institutes of Health (U.S.) , Rare Diseases/diagnosis , Syndrome , United States
7.
Am J Ophthalmol Case Rep ; 10: 244-248, 2018 Jun.
Article En | MEDLINE | ID: mdl-29780943

PURPOSE: We present the first detailed ophthalmic description of a child with Helsmoortel-Van der Aa Syndrome (HVDAS), including longitudinal follow-up and analysis. OBSERVATIONS: After extensive workup, a young child with poor visual behavior, hypotonic cerebral palsy, intellectual disability, and global developmental delay was found to have a heterozygous de novo mutation in the ADNP gene and diagnosed with HVDAS. Ophthalmic findings were remarkable for progressive nystagmus, macular pigment mottling, mild foveal hypoplasia with abnormal macular laminations, persistent rod dysfunction with electronegative waveform, and progressive cone degeneration. CONCLUSIONS AND IMPORTANCE: Patients with HVDAS are known to have abnormal visual behavior due to refractive or cortical impairment. However, we present the first description, to our knowledge, of an association with retinal mal-development and degeneration. Thus, patients with HVDAS should be referred for ophthalmic genetics evaluation, and HVDAS should be on the differential diagnosis for young children with global developmental delay who present with nystagmus, rod and cone dysfunction with electronegative waveform, and relative lack of severe structural degeneration on optical coherence tomography.

8.
Am J Hum Genet ; 102(3): 494-504, 2018 03 01.
Article En | MEDLINE | ID: mdl-29478781

ATP synthase, H+ transporting, mitochondrial F1 complex, δ subunit (ATP5F1D; formerly ATP5D) is a subunit of mitochondrial ATP synthase and plays an important role in coupling proton translocation and ATP production. Here, we describe two individuals, each with homozygous missense variants in ATP5F1D, who presented with episodic lethargy, metabolic acidosis, 3-methylglutaconic aciduria, and hyperammonemia. Subject 1, homozygous for c.245C>T (p.Pro82Leu), presented with recurrent metabolic decompensation starting in the neonatal period, and subject 2, homozygous for c.317T>G (p.Val106Gly), presented with acute encephalopathy in childhood. Cultured skin fibroblasts from these individuals exhibited impaired assembly of F1FO ATP synthase and subsequent reduced complex V activity. Cells from subject 1 also exhibited a significant decrease in mitochondrial cristae. Knockdown of Drosophila ATPsynδ, the ATP5F1D homolog, in developing eyes and brains caused a near complete loss of the fly head, a phenotype that was fully rescued by wild-type human ATP5F1D. In contrast, expression of the ATP5F1D c.245C>T and c.317T>G variants rescued the head-size phenotype but recapitulated the eye and antennae defects seen in other genetic models of mitochondrial oxidative phosphorylation deficiency. Our data establish c.245C>T (p.Pro82Leu) and c.317T>G (p.Val106Gly) in ATP5F1D as pathogenic variants leading to a Mendelian mitochondrial disease featuring episodic metabolic decompensation.


Alleles , Metabolic Diseases/genetics , Mitochondrial Proton-Translocating ATPases/genetics , Mutation/genetics , Protein Subunits/genetics , Amino Acid Sequence , Base Sequence , Child , Child, Preschool , Female , Humans , Infant , Infant, Newborn , Loss of Function Mutation/genetics , Male , Mitochondria/metabolism , Mitochondria/ultrastructure , Mitochondrial Proton-Translocating ATPases/chemistry , Protein Subunits/chemistry
9.
Epilepsia ; 58(10): 1771-1781, 2017 10.
Article En | MEDLINE | ID: mdl-28762469

OBJECTIVES: Glutaric acidemia type I (GA-I) is an inherited neurometabolic disorder caused by deficiency of glutaryl-CoA dehydrogenase (GCDH) and characterized by increased levels of glutaric, 3-OH-glutaric, and glutaconic acids in the brain parenchyma. The increment of these organic acids inhibits glutamate decarboxylase (GAD) and consequently lowers the γ-aminobutyric acid (GABA) synthesis. Untreated patients exhibit severe neurologic deficits during development, including epilepsy, especially following an acute encephalopathy outbreak. In this work, we evaluated the role of the GABAergic system on epileptogenesis in GA-I using the Gcdh-/- mice exposed to a high lysine diet (Gcdh-/- -Lys). METHODS: Spontaneous recurrent seizures (SRS), seizure susceptibility, and changes in brain oscillations were evaluated by video-electroencephalography (EEG). Cortical GABAergic synaptic transmission was evaluated using electrophysiologic and neurochemical approaches. RESULTS: SRS were observed in 72% of Gcdh-/- -Lys mice, whereas no seizures were detected in age-matched controls (Gcdh+/+ or Gcdh-/- receiving normal diet). The severity and number of PTZ-induced seizures were higher in Gcdh-/- -Lys mice. EEG spectral analysis showed a significant decrease in theta and gamma oscillations and predominant delta waves in Gcdh-/- -Lys mice, associated with increased EEG left index. Analysis of cortical synaptosomes revealed a significantly increased percentage of glutamate release and decreased GABA release in Gcdh-/- -Lys mice that were associated with a decrease in cortical GAD immunocontent and activity and confirmed by reduced frequency of inhibitory events in cortical pyramidal cells. SIGNIFICANCE: Using an experimental model with a phenotype similar to that of GA-I in humans-the Gcdh-/- mice under high lysine diet (Gcdh-/- -Lys)-we provide evidence that a reduction in cortical inhibition of Gcdh-/- -Lys mice, probably induced by GAD dysfunction, leads to hyperexcitability and increased slow oscillations associated with neurologic abnormalities in GA-I. Our findings offer a new perspective on the pathophysiology of brain damage in GA-I.


Amino Acid Metabolism, Inborn Errors/genetics , Brain Diseases, Metabolic/genetics , Brain/drug effects , Epilepsy/genetics , Glutaryl-CoA Dehydrogenase/deficiency , Glutaryl-CoA Dehydrogenase/genetics , gamma-Aminobutyric Acid/drug effects , Amino Acid Metabolism, Inborn Errors/metabolism , Animals , Blotting, Western , Brain Diseases, Metabolic/metabolism , Chromatography, High Pressure Liquid , Epilepsy/metabolism , GABA Antagonists/pharmacology , Glutamate Decarboxylase , Glutamic Acid/drug effects , Glutamic Acid/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Mice , Mice, Knockout , Pentylenetetrazole/pharmacology , Synaptosomes/drug effects , Synaptosomes/metabolism , gamma-Aminobutyric Acid/metabolism
11.
Am J Med Genet A ; 173(9): 2500-2504, 2017 Sep.
Article En | MEDLINE | ID: mdl-28657663

Pompe disease is a rare inherited metabolic disorder of glycogen metabolism caused by mutations in the GAA gene, encoding the acid α-1,4 glucosidase. Successful diagnosis of Pompe disease is achieved by clinical and biochemical evaluation followed by confirmation with DNA testing. Here, we report a male infant with a prenatal onset of cardiac symptoms and enzyme testing consistent with Pompe disease, but DNA testing by Sanger sequencing revealed no pathogenic variants. Due to the strong indication from clinical, enzymatic, and histological studies (despite the absence of molecular confirmation by traditional Sanger sequencing), enzyme replacement therapy (ERT) for Pompe disease was initiated. Reanalysis of the patient's DNA sample using next generation sequencing (NGS) of a panel of target genes causing glycogen storage disorders demonstrated compound heterozygosity for a point mutation and an exonic deletion in the GAA gene. This case illustrates the value of astute clinical judgement in patient management as well as the power of target capture deep NGS in the simultaneous detection of both a point mutation and a heterozygous exonic deletion by correcting pitfalls of the traditional PCR based sequencing, namely; allele dropout and the inability to detect exonic deletions.


Glycogen Storage Disease Type II/genetics , High-Throughput Nucleotide Sequencing/methods , Pathology, Molecular/methods , alpha-Glucosidases/genetics , Exons/genetics , Glycogen Storage Disease Type II/diagnosis , Glycogen Storage Disease Type II/physiopathology , Heterozygote , Humans , Infant , Male , Point Mutation/genetics
12.
J Inherit Metab Dis ; 40(1): 75-101, 2017 01.
Article En | MEDLINE | ID: mdl-27853989

Glutaric aciduria type I (GA-I; synonym, glutaric acidemia type I) is a rare inherited metabolic disease caused by deficiency of glutaryl-CoA dehydrogenase located in the catabolic pathways of L-lysine, L-hydroxylysine, and L-tryptophan. The enzymatic defect results in elevated concentrations of glutaric acid, 3-hydroxyglutaric acid, glutaconic acid, and glutaryl carnitine in body tissues, which can be reliably detected by gas chromatography/mass spectrometry (organic acids) and tandem mass spectrometry (acylcarnitines). Most untreated individuals with GA-I experience acute encephalopathic crises during the first 6 years of life that are triggered by infectious diseases, febrile reaction to vaccinations, and surgery. These crises result in striatal injury and consequent dystonic movement disorder; thus, significant mortality and morbidity results. In some patients, neurologic disease may also develop without clinically apparent crises at any age. Neonatal screening for GA-I us being used in a growing number of countries worldwide and is cost effective. Metabolic treatment, consisting of low lysine diet, carnitine supplementation, and intensified emergency treatment during catabolism, is effective treatment and improves neurologic outcome in those individuals diagnosed early; treatment after symptom onset, however, is less effective. Dietary treatment is relaxed after age 6 years and should be supervised by specialized metabolic centers. The major aim of this second revision of proposed recommendations is to re-evaluate the previous recommendations (Kölker et al. J Inherit Metab Dis 30:5-22, 2007b; J Inherit Metab Dis 34:677-694, 2011) and add new research findings, relevant clinical aspects, and the perspective of affected individuals.


Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/drug therapy , Brain Diseases, Metabolic/diagnosis , Brain Diseases, Metabolic/drug therapy , Glutaryl-CoA Dehydrogenase/deficiency , Amino Acid Metabolism, Inborn Errors/metabolism , Brain Diseases, Metabolic/metabolism , Dietary Supplements , Glutarates/metabolism , Glutaryl-CoA Dehydrogenase/metabolism , Humans , Lysine/metabolism
13.
Nutr Res ; 36(1): 101-8, 2016 Jan.
Article En | MEDLINE | ID: mdl-26773786

A higher incidence of osteopenia is observed among children with inherited metabolic disorders (inborn errors of metabolism, or IEMs) who consume medical food-based diets that restrict natural vitamin D-containing food sources. We evaluated the vitamin D status of children with IEMs who live in the Pacific Northwest with limited sun exposure and determined whether bone mineral density (BMD) in children with phenylketonuria (PKU), the most common IEM, correlated with diet or biochemical markers of bone metabolism. We hypothesized that children with IEMs would have lower serum vitamin D concentrations than controls and that some children with PKU would have reduced bone mineralization. A retrospective record review of 88 patients with IEMs, and 445 children on unrestricted diets (controls) found the 25-hydroxyvitamin D concentrations were normal and not significantly different between groups (IEM patients, 27.1 ± 10.9; controls, 27.6 ± 11.2). Normal BMD at the hip or spine (-2

25-Hydroxyvitamin D 2/blood , Bone Density , Bone Diseases, Developmental/prevention & control , Calcifediol/blood , Food, Formulated , Metabolism, Inborn Errors/diet therapy , Vitamin D Deficiency/prevention & control , Academic Medical Centers , Adolescent , Adult , Biomarkers/blood , Bone Diseases, Developmental/epidemiology , Bone Diseases, Developmental/etiology , Child , Cohort Studies , Cross-Sectional Studies , Electronic Health Records , Food, Formulated/adverse effects , Humans , Incidence , Metabolism, Inborn Errors/blood , Metabolism, Inborn Errors/physiopathology , Oregon/epidemiology , Phenylketonurias/blood , Phenylketonurias/diet therapy , Phenylketonurias/physiopathology , Retrospective Studies , Risk , Vitamin D Deficiency/epidemiology , Vitamin D Deficiency/etiology , Young Adult
14.
Parasit Vectors ; 9: 7, 2016 Jan 05.
Article En | MEDLINE | ID: mdl-26728034

BACKGROUND: Mitochondria play essential biological functions including the synthesis and trafficking of porphyrins and iron/sulfur clusters (ISC), processes that in mammals involve the mitochondrial ATP-Binding Cassette (ABC) transporters ABCB6 and ABCB7, respectively. The mitochondrion of pathogenic protozoan parasites such as Leishmania is a promising goal for new therapeutic approaches. Leishmania infects human macrophages producing the neglected tropical disease known as leishmaniasis. Like most trypanosomatid parasites, Leishmania is auxotrophous for heme and must acquire porphyrins from the host. METHODS: LmABCB3, a new Leishmania major protein with significant sequence similarity to human ABCB6/ABCB7, was identified and characterized using bioinformatic tools. Fluorescent microscopy was used to determine its cellular localization, and its level of expression was modulated by molecular genetic techniques. Intracellular in vitro assays were used to demonstrate its role in amastigotes replication, and an in vivo mouse model was used to analyze its role in virulence. Functional characterization of LmABCB3 was carried out in Leishmania promastigotes and Saccharomyces cerevisiae. Structural analysis of LmABCB3 was performed using molecular modeling software. RESULTS: LmABCB3 is an atypical ABC half-transporter that has a unique N-terminal extension not found in any other known ABC protein. This extension is required to target LmABCB3 to the mitochondrion and includes a potential metal-binding domain. We have shown that LmABCB3 interacts with porphyrins and is required for the mitochondrial synthesis of heme from a host precursor. We also present data supporting a role for LmABCB3 in the biogenesis of cytosolic ISC, essential cofactors for cell viability in all three kingdoms of life. LmABCB3 fully complemented the severe growth defect shown in yeast lacking ATM1, an orthologue of human ABCB7 involved in exporting from the mitochondria a gluthatione-containing compound required for the generation of cytosolic ISC. Indeed, docking analyzes performed with a LmABCB3 structural model using trypanothione, the main thiol in this parasite, as a ligand showed how both, LmABCB3 and yeast ATM1, contain a similar thiol-binding pocket. Additionally, we show solid evidence suggesting that LmABCB3 is an essential gene as dominant negative inhibition of LmABCB3 is lethal for the parasite. Moreover, the abrogation of only one allele of the gene did not impede promastigote growth in axenic culture but prevented the replication of intracellular amastigotes and the virulence of the parasites in a mouse model of cutaneous leishmaniasis. CONCLUSIONS: Altogether our results present the previously undescribed LmABCB3 as an unusual mitochondrial ABC transporter essential for Leishmania survival through its role in the generation of heme and cytosolic ISC. Hence, LmABCB3 could represent a novel target to combat leishmaniasis.


ATP-Binding Cassette Transporters/metabolism , Leishmania major/genetics , Leishmaniasis/parasitology , ATP-Binding Cassette Transporters/genetics , Animals , Heme/metabolism , Humans , Iron/metabolism , Leishmania major/metabolism , Leishmania major/pathogenicity , Male , Mice , Mice, Inbred C57BL , Mitochondria/metabolism , Models, Molecular , Protein Transport , Sulfur/metabolism , Virulence
15.
Genet Med ; 18(9): 933-9, 2016 09.
Article En | MEDLINE | ID: mdl-26820065

PURPOSE: Infant mortality in Alaska is highest among Alaska Native people from western/northern Alaska, a population with a high prevalence of a genetic variant (c.1436C>T; the arctic variant) of carnitine palmitoyltransferase 1A (CPT1A). METHODS: We performed an unmatched case-control study to determine the relationship between the arctic variant and infant mortality. The cases were 110 Alaska Native infant deaths from 2006 to 2010 and the controls were 395 Alaska Native births from the same time period. In addition to the overall analysis, we conducted two subanalyses, one limited to subjects from western/northern Alaska and one limited to infants heterozygous or homozygous for the arctic variant. RESULTS: Among western/northern Alaska residents, 66% of cases and 61% of controls were homozygous (adjusted odds ratio (aOR): 2.5; 95% confidence interval (CI): 1.3, 5.0). Among homozygous or heterozygous infants, 58% of cases and 44% of controls were homozygous (aOR: 2.3; 95% CI: 1.3, 4.0). Deaths associated with infection were more likely to be homozygous (OR: 2.9; 95% CI: 1.0-8.0). Homozygosity was strongly associated with a premorbid history of pneumonia, sepsis, or meningitis. CONCLUSION: Homozygosity for the arctic variant is associated with increased risk of infant mortality, which may be mediated in part by an increase in infectious disease risk. Further studies are needed to determine whether the association we report represents a causal association between the CPT1A arctic variant and infectious disease-specific mortality.Genet Med 18 9, 933-939.


Carnitine O-Palmitoyltransferase/genetics , Communicable Diseases/genetics , Infant Mortality , Neonatal Screening , Alaska , Alaskan Natives/genetics , Communicable Diseases/mortality , Communicable Diseases/pathology , Female , Genetic Association Studies , Genetic Variation , Homozygote , Humans , Indians, North American , Infant , Infant, Newborn , Male , Meningitis/genetics , Meningitis/mortality , Pneumonia/genetics , Pneumonia/mortality , Risk Factors , Sepsis/genetics , Sepsis/mortality
16.
Mol Genet Metab ; 116(4): 252-9, 2015 Dec.
Article En | MEDLINE | ID: mdl-26490222

BACKGROUND: Arginine:glycine aminotransferase (AGAT) (GATM) deficiency is an autosomal recessive inborn error of creative synthesis. OBJECTIVE: We performed an international survey among physicians known to treat patients with AGAT deficiency, to assess clinical characteristics and long-term outcomes of this ultra-rare condition. RESULTS: 16 patients from 8 families of 8 different ethnic backgrounds were included. 1 patient was asymptomatic when diagnosed at age 3 weeks. 15 patients diagnosed between 16 months and 25 years of life had intellectual disability/developmental delay (IDD). 8 patients also had myopathy/proximal muscle weakness. Common biochemical denominators were low/undetectable guanidinoacetate (GAA) concentrations in urine and plasma, and low/undetectable cerebral creatine levels. 3 families had protein truncation/null mutations. The rest had missense and splice mutations. Treatment with creatine monohydrate (100-800 mg/kg/day) resulted in almost complete restoration of brain creatine levels and significant improvement of myopathy. The 2 patients treated since age 4 and 16 months had normal cognitive and behavioral development at age 10 and 11 years. Late treated patients had limited improvement of cognitive functions. CONCLUSION: AGAT deficiency is a treatable intellectual disability. Early diagnosis may prevent IDD and myopathy. Patients with unexplained IDD with and without myopathy should be assessed for AGAT deficiency by determination of urine/plasma GAA and cerebral creatine levels (via brain MRS), and by GATM gene sequencing.


Amidinotransferases/deficiency , Amino Acid Metabolism, Inborn Errors/drug therapy , Creatine/therapeutic use , Intellectual Disability/drug therapy , Muscular Diseases/drug therapy , Speech Disorders/drug therapy , Adolescent , Amidinotransferases/chemistry , Amidinotransferases/genetics , Amino Acid Metabolism, Inborn Errors/diagnosis , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/physiopathology , Child , Child, Preschool , Creatine/deficiency , Developmental Disabilities/diagnosis , Developmental Disabilities/drug therapy , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Gene Expression , Genes, Recessive , Glycine/analogs & derivatives , Glycine/blood , Glycine/deficiency , Glycine/urine , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Intellectual Disability/physiopathology , Magnetic Resonance Spectroscopy , Male , Models, Molecular , Muscular Diseases/diagnosis , Muscular Diseases/genetics , Muscular Diseases/physiopathology , Mutation , Protein Structure, Secondary , Protein Structure, Tertiary , Sequence Analysis, DNA , Speech Disorders/diagnosis , Speech Disorders/genetics , Speech Disorders/physiopathology , Treatment Outcome , Young Adult
17.
J Neurol Sci ; 344(1-2): 105-13, 2014 Sep 15.
Article En | MEDLINE | ID: mdl-24996493

We evaluated the antioxidant defense system and protein oxidative damage in the brain and liver of 15-day-old GCDH deficient knockout (Gcdh(-/-)) mice following an acute intraperitoneal administration of Lys (8 µmol/g). We determined reduced glutathione (GSH) concentrations, sulfhydryl content, carbonyl formation and the activities of the antioxidant enzymes glutathione peroxidase (GPx), superoxide dismutase (SOD), catalase (CAT) and glutathione reductase (GR) in the brain and liver of these animals. 2',7'-dihydrodichlorofluorescein (DCFH) oxidation was also measured as an index of free radical formation. The only parameters altered in Gcdh(-/-) compared to wild type (Gcdh(+/+)) mice were a reduction of liver GSH concentrations and of brain sulfhydryl content. Acute Lys injection provoked a decrease of GSH concentration in the brain and sulfhydryl content in the liver, and an increase in carbonyl formation in the brain and liver of Gcdh(-/-) mice. Lys administration also induced a decrease of all antioxidant enzyme activities in the brain, as well as an increase of the activities of SOD and CAT in the liver of Gcdh(-/-) mice. Finally, Lys elicited a marked increase of DCFH oxidation in the brain and liver. It is concluded that Lys overload compromises the brain antioxidant defenses and induces protein oxidation probably secondary to reactive species generation in infant Gcdh(+/+) mice.


Amino Acid Metabolism, Inborn Errors/pathology , Brain Diseases, Metabolic/pathology , Brain/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Lysine/therapeutic use , Neuroprotective Agents/therapeutic use , Oxidative Stress/drug effects , Amino Acid Metabolism, Inborn Errors/complications , Analysis of Variance , Animals , Animals, Newborn , Brain Diseases, Metabolic/complications , Brain Injuries/etiology , Brain Injuries/prevention & control , Catalase , Disease Models, Animal , Glutaryl-CoA Dehydrogenase/drug effects , Glutaryl-CoA Dehydrogenase/metabolism , Glutathione/metabolism , Glutathione Peroxidase , Lysine/pharmacology , Mice , Mice, Transgenic , Neuroprotective Agents/pharmacology , Oxidation-Reduction , Superoxide Dismutase
20.
PLoS One ; 9(3): e90477, 2014.
Article En | MEDLINE | ID: mdl-24594605

We determined mRNA expression of the ionotropic glutamate receptors NMDA (NR1, NR2A and NR2B subunits), AMPA (GluR2 subunit) and kainate (GluR6 subunit), as well as of the glutamate transporters GLAST and GLT1 in cerebral cortex and striatum of wild type (WT) and glutaryl-CoA dehydrogenase deficient (Gchh-/-) mice aged 7, 30 and 60 days. The protein expression levels of some of these membrane proteins were also measured. Overexpression of NR2A and NR2B in striatum and of GluR2 and GluR6 in cerebral cortex was observed in 7-day-old Gcdh-/-. There was also an increase of mRNA expression of all NMDA subunits in cerebral cortex and of NR2A and NR2B in striatum of 30-day-old Gcdh-/- mice. At 60 days of life, all ionotropic receptors were overexpressed in cerebral cortex and striatum of Gcdh-/- mice. Higher expression of GLAST and GLT1 transporters was also verified in cerebral cortex and striatum of Gcdh-/- mice aged 30 and 60 days, whereas at 7 days of life GLAST was overexpressed only in striatum from this mutant mice. Furthermore, high lysine intake induced mRNA overexpression of NR2A, NR2B and GLAST transcripts in striatum, as well as of GluR2 and GluR6 in both striatum and cerebral cortex of Gcdh-/- mice. Finally, we found that the protein expression of NR2A, NR2B, GLT1 and GLAST were significantly greater in cerebral cortex of Gcdh-/- mice, whereas NR2B and GLT1 was similarly enhanced in striatum, implying that these transcripts were translated into their products. These results provide evidence that glutamate receptor and transporter expression is higher in Gcdh-/- mice and that these alterations may be involved in the pathophysiology of GA I and possibly explain, at least in part, the vulnerability of striatum and cerebral cortex to injury in patients affected by GA I.


Amino Acid Metabolism, Inborn Errors/pathology , Amino Acid Transport System X-AG/metabolism , Brain Diseases, Metabolic/pathology , Cerebral Cortex/metabolism , Glutaryl-CoA Dehydrogenase/deficiency , Neostriatum/metabolism , Receptors, Glutamate/metabolism , Amino Acid Metabolism, Inborn Errors/enzymology , Amino Acid Transport System X-AG/genetics , Animals , Brain Diseases, Metabolic/enzymology , Cerebral Cortex/pathology , Diet , Female , Gene Expression Regulation , Glutaryl-CoA Dehydrogenase/metabolism , Lysine/metabolism , Male , Mice , Neostriatum/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Glutamate/genetics
...