Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
2.
Genet Med ; 25(6): 100314, 2023 06.
Article in English | MEDLINE | ID: mdl-36305855

ABSTRACT

PURPOSE: This study aimed to define the genotypic and phenotypic spectrum of reversible acute liver failure (ALF) of infancy resulting from biallelic pathogenic TRMU variants and determine the role of cysteine supplementation in its treatment. METHODS: Individuals with biallelic (likely) pathogenic variants in TRMU were studied within an international retrospective collection of de-identified patient data. RESULTS: In 62 individuals, including 30 previously unreported cases, we described 47 (likely) pathogenic TRMU variants, of which 17 were novel, and 1 intragenic deletion. Of these 62 individuals, 42 were alive at a median age of 6.8 (0.6-22) years after a median follow-up of 3.6 (0.1-22) years. The most frequent finding, occurring in all but 2 individuals, was liver involvement. ALF occurred only in the first year of life and was reported in 43 of 62 individuals; 11 of whom received liver transplantation. Loss-of-function TRMU variants were associated with poor survival. Supplementation with at least 1 cysteine source, typically N-acetylcysteine, improved survival significantly. Neurodevelopmental delay was observed in 11 individuals and persisted in 4 of the survivors, but we were unable to determine whether this was a primary or a secondary consequence of TRMU deficiency. CONCLUSION: In most patients, TRMU-associated ALF was a transient, reversible disease and cysteine supplementation improved survival.


Subject(s)
Liver Failure, Acute , Liver Failure , Adolescent , Child , Child, Preschool , Humans , Infant , Young Adult , Acetylcysteine/therapeutic use , Liver Failure/drug therapy , Liver Failure/genetics , Liver Failure, Acute/drug therapy , Liver Failure, Acute/genetics , Mitochondrial Proteins/genetics , Mutation , Retrospective Studies , tRNA Methyltransferases/genetics
3.
J Pediatr ; 249: 50-58.e2, 2022 10.
Article in English | MEDLINE | ID: mdl-35709957

ABSTRACT

OBJECTIVE: To characterize the longitudinal natural history of disease progression in pediatric subjects affected with mucopolysaccharidosis (MPS) IIIB. STUDY DESIGN: Sixty-five children with a confirmed diagnosis of MPS IIIB were enrolled into 1 of 2 natural history studies and followed for up to 4 years. Cognitive and adaptive behavior functions were analyzed in all subjects, and volumetric magnetic resonance imaging analysis of liver, spleen, and brain, as well as levels of heparan sulfate (HS) and heparan sulfate nonreducing ends (HS-NRE), were measured in a subset of subjects. RESULTS: The majority of subjects with MPS IIIB achieved an apex on both cognition and adaptive behavior age equivalent scales between age 3 and 6 years. Development quotients for both cognition and adaptive behavior follow a linear trajectory by which subjects reach a nadir with a score <25 for an age equivalent of 24 months by age 8 years on average and by 13.5 years at the latest. All tested subjects (n = 22) had HS and HS-NRE levels above the normal range in cerebrospinal fluid and plasma, along with signs of hepatomegaly. Subjects lost an average of 26 mL of brain volume (-2.7%) over 48 weeks, owing entirely to a loss of cortical gray matter (32 mL; -6.5%). CONCLUSIONS: MPS IIIB exists along a continuum based on cognitive decline and cortical gray matter atrophy. Although a few individuals with MPS IIIB have an attenuated phenotype, the majority follow predicted trajectories for both cognition and adaptive behavior. TRIAL REGISTRATION: ClinicalTrials.gov identifiers NCT02493998, NCT03227042, and NCT02754076.


Subject(s)
Mucopolysaccharidosis III , Atrophy/pathology , Brain/diagnostic imaging , Brain/pathology , Gray Matter , Heparitin Sulfate , Humans , Magnetic Resonance Imaging , Mucopolysaccharidosis III/diagnosis
4.
Blood ; 139(12): 1833-1849, 2022 03 24.
Article in English | MEDLINE | ID: mdl-35081253

ABSTRACT

Niemann-Pick disease type C1 (NP-C1) is a rare lysosomal storage disorder resulting from mutations in an endolysosomal cholesterol transporter, NPC1. Despite typically presenting with pronounced neurological manifestations, NP-C1 also resembles long-term congenital immunodeficiencies that arise from impairment of cytotoxic T lymphocyte (CTL) effector function. CTLs kill their targets through exocytosis of the contents of lysosome-like secretory cytotoxic granules (CGs) that store and ultimately release the essential pore-forming protein perforin and proapoptotic serine proteases, granzymes, into the synapse formed between the CTL and target cell. We discovered that NPC1 deficiency increases CG lipid burden, impairs autophagic flux through stalled trafficking of the transcription factor EB (TFEB), and dramatically reduces CTL cytotoxicity. Using a variety of immunological and cell biological techniques, we found that the cytotoxic defect arises specifically from impaired perforin pore formation. We demonstrated defects of CTL function of varying severity in patients with NP-C1, with the greatest losses of function associated with the most florid and/or earliest disease presentations. Remarkably, perforin function and CTL cytotoxicity were restored in vitro by promoting lipid clearance with therapeutic 2-hydroxypropyl-ß-cyclodextrin; however, restoration of autophagy through TFEB overexpression was ineffective. Overall, our study revealed that NPC1 deficiency has a deleterious impact on CTL (but not natural killer cell) cytotoxicity that, in the long term, may predispose patients with NP-C1 to atypical infections and impaired immune surveillance more generally.


Subject(s)
Niemann-Pick Disease, Type A , Niemann-Pick Disease, Type C , Cholesterol/metabolism , Granzymes , Humans , Niemann-Pick Disease, Type C/metabolism , Perforin/genetics , T-Lymphocytes, Cytotoxic/metabolism
5.
Int J Mol Sci ; 23(2)2022 Jan 17.
Article in English | MEDLINE | ID: mdl-35055180

ABSTRACT

Pyridine Nucleotide-Disulfide Oxidoreductase Domain 2 (PYROXD2; previously called YueF) is a mitochondrial inner membrane/matrix-residing protein and is reported to regulate mitochondrial function. The clinical importance of PYROXD2 has been unclear, and little is known of the protein's precise biological function. In the present paper, we report biallelic variants in PYROXD2 identified by genome sequencing in a patient with suspected mitochondrial disease. The child presented with acute neurological deterioration, unresponsive episodes, and extreme metabolic acidosis, and received rapid genomic testing. He died shortly after. Magnetic resonance imaging (MRI) brain imaging showed changes resembling Leigh syndrome, one of the more common childhood mitochondrial neurological diseases. Functional studies in patient fibroblasts showed a heightened sensitivity to mitochondrial metabolic stress and increased mitochondrial superoxide levels. Quantitative proteomic analysis demonstrated decreased levels of subunits of the mitochondrial respiratory chain complex I, and both the small and large subunits of the mitochondrial ribosome, suggesting a mitoribosomal defect. Our findings support the critical role of PYROXD2 in human cells, and suggest that the biallelic PYROXD2 variants are associated with mitochondrial dysfunction, and can plausibly explain the child's clinical presentation.


Subject(s)
Leigh Disease/diagnostic imaging , Mutation, Missense , Tumor Suppressor Proteins/genetics , Fatal Outcome , Humans , Infant , Leigh Disease/genetics , Magnetic Resonance Imaging , Male , Mitochondrial Proteins/metabolism , Models, Molecular , Proteomics , Sequence Analysis, RNA , Tumor Suppressor Proteins/chemistry , Whole Genome Sequencing
6.
Am J Hum Genet ; 108(11): 2130-2144, 2021 11 04.
Article in English | MEDLINE | ID: mdl-34653363

ABSTRACT

Congenital disorders of glycosylation (CDGs) form a group of rare diseases characterized by hypoglycosylation. We here report the identification of 16 individuals from nine families who have either inherited or de novo heterozygous missense variants in STT3A, leading to an autosomal-dominant CDG. STT3A encodes the catalytic subunit of the STT3A-containing oligosaccharyltransferase (OST) complex, essential for protein N-glycosylation. Affected individuals presented with variable skeletal anomalies, short stature, macrocephaly, and dysmorphic features; half had intellectual disability. Additional features included increased muscle tone and muscle cramps. Modeling of the variants in the 3D structure of the OST complex indicated that all variants are located in the catalytic site of STT3A, suggesting a direct mechanistic link to the transfer of oligosaccharides onto nascent glycoproteins. Indeed, expression of STT3A at mRNA and steady-state protein level in fibroblasts was normal, while glycosylation was abnormal. In S. cerevisiae, expression of STT3 containing variants homologous to those in affected individuals induced defective glycosylation of carboxypeptidase Y in a wild-type yeast strain and expression of the same mutants in the STT3 hypomorphic stt3-7 yeast strain worsened the already observed glycosylation defect. These data support a dominant pathomechanism underlying the glycosylation defect. Recessive mutations in STT3A have previously been described to lead to a CDG. We present here a dominant form of STT3A-CDG that, because of the presence of abnormal transferrin glycoforms, is unusual among dominant type I CDGs.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Genes, Dominant , Hexosyltransferases/genetics , Membrane Proteins/genetics , Musculoskeletal Diseases/genetics , Nervous System Diseases/genetics , Adolescent , Adult , Amino Acid Sequence , Catalytic Domain , Child, Preschool , Female , Heterozygote , Hexosyltransferases/chemistry , Humans , Male , Membrane Proteins/chemistry , Middle Aged , Pedigree , Sequence Homology, Amino Acid
7.
Am J Med Genet A ; 185(3): 909-915, 2021 03.
Article in English | MEDLINE | ID: mdl-33369132

ABSTRACT

We describe 10 females with ornithine transcarbamylase (OTC) deficiency and liver dysfunction, revealing a unique pattern of hepatocyte injury in which initial hyperammonemia and coagulopathy is followed by a delayed peak in aminotransferase levels. None of the patients required urgent liver transplantation, though five eventually underwent transplant for recurrent metabolic crises. We intend that this novel observation will initiate further investigations into the pathophysiology of liver dysfunction in OTC-deficient patients, and ultimately lead to the development of therapies and prevent the need for liver transplant.


Subject(s)
Alanine Transaminase/blood , Liver Diseases/etiology , Ornithine Carbamoyltransferase Deficiency Disease/complications , Age of Onset , Amino Acid Substitution , Aspartate Aminotransferases/blood , Biomarkers , Child, Preschool , Combined Modality Therapy , Developmental Disabilities/genetics , Disease Progression , Female , Hemorrhagic Disorders/etiology , Humans , Hyperammonemia/genetics , Infant , International Normalized Ratio , Liver Diseases/blood , Liver Diseases/surgery , Liver Transplantation , Mutation, Missense , Ornithine Carbamoyltransferase Deficiency Disease/blood , Ornithine Carbamoyltransferase Deficiency Disease/diet therapy , Ornithine Carbamoyltransferase Deficiency Disease/surgery , Vomiting/genetics
8.
Intern Med J ; 50 Suppl 4: 5-27, 2020 11.
Article in English | MEDLINE | ID: mdl-33210402

ABSTRACT

Lysosomal storage diseases (LSD) comprise a rare and heterogeneous group of nearly 50 heritable metabolic disorders caused by mutations in proteins critical for cellular lysosomal function. Defects in the activity of these proteins in multiple organs leads to progressive intra-lysosomal accumulation of specific substrates, resulting in disruption of cellular functions, extracellular inflammatory responses, tissue damage and organ dysfunction. The classification and clinical presentation of different LSD are dependent on the type of accumulated substrate. Some clinical signs and symptoms are common across multiple LSD, while others are more specific to a particular syndrome. Due to the rarity and wide clinical diversity of LSD, identification and diagnosis can be challenging, and in many cases diagnosis is delayed for months or years. Treatments, such as enzyme replacement therapy, haemopoietic stem cell transplantation and substrate reduction therapy, are now available for some of the LSD. For maximum effect, therapy must be initiated prior to the occurrence of irreversible tissue damage, highlighting the importance of prompt diagnosis. Herein, we discuss the clinical presentation, diagnosis and treatment of four of the treatable LSD: Gaucher disease, Fabry disease, Pompe disease, and two of the mucopolysaccharidoses (I and II). For each disease, we present illustrative case studies to help increase awareness of their clinical presentation and possible treatment outcomes.


Subject(s)
Enzyme Replacement Therapy , Fabry Disease/therapy , Gaucher Disease/therapy , Glycogen Storage Disease Type II/therapy , Lysosomal Storage Diseases/therapy , Mucopolysaccharidosis II/therapy , Mucopolysaccharidosis I/therapy , Adult , Child, Preschool , Fabry Disease/diagnosis , Female , Gaucher Disease/diagnosis , Glycogen Storage Disease Type II/diagnosis , Hematopoietic Stem Cell Transplantation , Humans , Lysosomal Storage Diseases/drug therapy , Male , Mucopolysaccharidosis I/diagnosis , Mucopolysaccharidosis II/diagnosis
9.
J. inborn errors metab. screen ; 8: e20200001, 2020. tab, graf
Article in English | LILACS-Express | LILACS | ID: biblio-1135005

ABSTRACT

Abstract The safety and efficacy of elosulfase alfa were evaluated in a multicenter, open-label, phase 3b study in Australian Morquio A patients, consisting of a 49-week initial phase and an extension phase until elosulfase alfa was government funded. Thirteen patients (1-27 years) were enrolled. No new safety concerns were identified over 138 weeks. Most drug-related adverse events were mild or moderate in severity; none led to study discontinuation. After 49 weeks of treatment, median improvements from baseline were seen in the 6-minute walk test (+41.0 m), 3-minute stair climb test (+14.0 stairs/min), forced vital capacity (+16.4%), forced expiratory volume in 1 second (+14.1%), urine keratan sulfate (-7.1 µg/mg creatinine), and pain intensity. Growth, cardiac function, sleep, and quality of life results were mixed or stable. These results provide further evidence of the acceptable safety/tolerability profile of elosulfase alfa. The improvements in endurance, pulmonary function, and pain support findings from previous studies.

11.
Semin Pediatr Neurol ; 26: 2-9, 2018 07.
Article in English | MEDLINE | ID: mdl-29961509

ABSTRACT

Brown-Vialetto-van Laere syndrome is characterized by a progressive sensorimotor neuropathy, optic atrophy, hearing loss, bulbar dysfunction, and respiratory insufficiency. Mutations in SLC52A2 and SLC52A3, encoding riboflavin transporters RFVT2 and RFVT3, respectively, are the genetic basis of this disorder, often referred to as riboflavin transporter deficiency types 2 and 3, respectively. We present cases of both types of riboflavin transporter deficiency, highlighting the distinguishing clinical features of a rapidly progressive motor or sensorimotor axonal neuropathy, optic atrophy, sensorineural hearing loss, and bulbar dysfunction. One child presented with isolated central apnea and hypoventilation, not previously described in genetically confirmed Brown-Vialetto-van Laere, later complicated by diaphragmatic paralysis secondary to phrenic nerve palsy. Magnetic resonance imaging showed T2 hyperintensity in the dorsal spinal cord in 2 children, as well as previously unreported cervical nerve root enlargement and cauda equina ventral nerve root enhancement in 1 child. Novel homozygous mutations were identified in each gene-a NM_024531.4(SLC52A2):c.505C > T, NP_078807.1(SLC52A2):p.(Arg169Cys) variant in SLC52A2 and NM_033409.3(SLC52A3):c.1316G > A, NP_212134.3(SLC52A3):p.(Gly439Asp) variant in SLC52A3. Both treated children showed improvement on high-dose riboflavin supplementation, highlighting the importance of early recognition of this treatable clinical entity.


Subject(s)
Bulbar Palsy, Progressive/diagnostic imaging , Bulbar Palsy, Progressive/genetics , Hearing Loss, Sensorineural/diagnostic imaging , Hearing Loss, Sensorineural/genetics , Brain/diagnostic imaging , Bulbar Palsy, Progressive/physiopathology , Bulbar Palsy, Progressive/therapy , Child, Preschool , Consanguinity , Female , Hearing Loss, Sensorineural/physiopathology , Hearing Loss, Sensorineural/therapy , Humans , Infant , Male , Membrane Transport Proteins/genetics , Receptors, G-Protein-Coupled/genetics , Spinal Cord/diagnostic imaging
12.
Cogn Neuropsychol ; 34(6): 347-356, 2017 09.
Article in English | MEDLINE | ID: mdl-29210605

ABSTRACT

Mucopolysaccharidosis Type II (MPS II) or Hunter Syndrome is a rare X-linked condition, due to a defect in a lysosomal enzyme involved in the breakdown of glycosaminoglycans. It is a progressive condition with worsening over time; however, symptom severity and progression rates vary. Normal intellectual function has been reported in males with mild MPS II but few studies are available that provide comprehensive cognitive profiles. Enzyme replacement therapy (ERT) can stabilize physical symptoms and has become standard treatment. Whether ERT can influence cognition is currently unknown. Considering this, we conducted cognitive, fine motor, and behavioural assessments with three males (7;6-12;1 years) with mild MPS II before and after ERT. Generally, cognition, fine motor skills, and behaviour were in the normal range; however, specific deficits in attention and executive function were identified. Following ERT, some memory improvements were seen. Executive deficits remained, and processing speed declined over time.


Subject(s)
Attention , Cognition , Executive Function , Mucopolysaccharidosis II/psychology , Child , Disease Progression , Enzyme Replacement Therapy , Humans , Iduronate Sulfatase/metabolism , Infant , Male , Memory , Mucopolysaccharidosis II/enzymology , Mucopolysaccharidosis II/therapy
13.
JAMA Pediatr ; 171(9): 855-862, 2017 09 01.
Article in English | MEDLINE | ID: mdl-28759686

ABSTRACT

Importance: Optimal use of whole-exome sequencing (WES) in the pediatric setting requires an understanding of who should be considered for testing and when it should be performed to maximize clinical utility and cost-effectiveness. Objectives: To investigate the impact of WES in sequencing-naive children suspected of having a monogenic disorder and evaluate its cost-effectiveness if WES had been available at different time points in their diagnostic trajectory. Design, Setting, and Participants: This prospective study was part of the Melbourne Genomics Health Alliance demonstration project. At the ambulatory outpatient clinics of the Victorian Clinical Genetics Services at the Royal Children's Hospital, Melbourne, Australia, children older than 2 years suspected of having a monogenic disorder were prospectively recruited from May 1 through November 30, 2015, by clinical geneticists after referral from general and subspecialist pediatricians. All children had nondiagnostic microarrays and no prior single-gene or panel sequencing. Exposures: All children underwent singleton WES with targeted phenotype-driven analysis. Main Outcomes and Measures: The study examined the clinical utility of a molecular diagnosis and the cost-effectiveness of alternative diagnostic trajectories, depending on timing of WES. Results: Of 61 children originally assessed, 44 (21 [48%] male and 23 [52%] female) aged 2 to 18 years (mean age at initial presentation, 28 months; range, 0-121 months) were recruited, and a diagnosis was achieved in 23 (52%) by singleton WES. The diagnoses were unexpected in 8 of 23 (35%), and clinical management was altered in 6 of 23 (26%). The mean duration of the diagnostic odyssey was 6 years, with each child having a mean of 19 tests and 4 clinical genetics and 4 nongenetics specialist consultations, and 26 (59%) underwent a procedure while under general anesthetic for diagnostic purposes. Economic analyses of the diagnostic trajectory identified that WES performed at initial tertiary presentation resulted in an incremental cost savings of A$9020 (US$6838) per additional diagnosis (95% CI, A$4304-A$15 404 [US$3263-US$11 678]) compared with the standard diagnostic pathway. Even if WES were performed at the first genetics appointment, there would be an incremental cost savings of A$5461 (US$4140) (95% CI, A$1433-A$10 557 [US$1086- US$8004]) per additional diagnosis compared with the standard diagnostic pathway. Conclusions and Relevance: Singleton WES in children with suspected monogenic conditions has high diagnostic yield, and cost-effectiveness is maximized by early application in the diagnostic pathway. Pediatricians should consider early referral of children with undiagnosed syndromes to clinical geneticists.


Subject(s)
Genetic Diseases, Inborn/diagnosis , Sequence Analysis, DNA/methods , Australia , Child , Child, Preschool , Cost-Benefit Analysis , Exome/genetics , Female , Genetic Diseases, Inborn/economics , Humans , Male , Mutation , Prospective Studies , Sequence Analysis, DNA/economics
14.
J Biol Chem ; 291(39): 20563-73, 2016 09 23.
Article in English | MEDLINE | ID: mdl-27519416

ABSTRACT

Methylmalonic aciduria (MMAuria), caused by deficiency of methylmalonyl-CoA mutase (MUT), usually presents in the newborn period with failure to thrive and metabolic crisis leading to coma or even death. Survivors remain at risk of metabolic decompensations and severe long term complications, notably renal failure and neurological impairment. We generated clinically relevant mouse models of MMAuria using a constitutive Mut knock-in (KI) allele based on the p.Met700Lys patient mutation, used homozygously (KI/KI) or combined with a knockout allele (KO/KI), to study biochemical and clinical MMAuria disease aspects. Transgenic Mut(ki/ki) and Mut(ko/ki) mice survive post-weaning, show failure to thrive, and show increased methylmalonic acid, propionylcarnitine, odd chain fatty acids, and sphingoid bases, a new potential biomarker of MMAuria. Consistent with genetic dosage, Mut(ko/ki) mice have lower Mut activity, are smaller, and show higher metabolite levels than Mut(ki/ki) mice. Further, Mut(ko/ki) mice exhibit manifestations of kidney and brain damage, including increased plasma urea, impaired diuresis, elevated biomarkers, and changes in brain weight. On a high protein diet, mutant mice display disease exacerbation, including elevated blood ammonia, and catastrophic weight loss, which, in Mut(ki/ki) mice, is rescued by hydroxocobalamin treatment. This study expands knowledge of MMAuria, introduces the discovery of new biomarkers, and constitutes the first in vivo proof of principle of cobalamin treatment in mut-type MMAuria.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Gene Dosage , Methylmalonyl-CoA Mutase , Phenotype , Quantitative Trait, Heritable , Amino Acid Metabolism, Inborn Errors/blood , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Ammonia/metabolism , Animals , Biomarkers/blood , Brain/metabolism , Brain/pathology , Carnitine/analogs & derivatives , Carnitine/blood , Dietary Proteins/adverse effects , Dietary Proteins/pharmacology , Disease Models, Animal , Gene Knock-In Techniques , Kidney/metabolism , Kidney/pathology , Methylmalonic Acid/blood , Methylmalonyl-CoA Mutase/genetics , Methylmalonyl-CoA Mutase/metabolism , Mice , Mice, Knockout
15.
Hum Mutat ; 37(7): 653-60, 2016 07.
Article in English | MEDLINE | ID: mdl-26931382

ABSTRACT

Congenital disorders of glycosylation (CDG) arise from pathogenic mutations in over 100 genes leading to impaired protein or lipid glycosylation. ALG1 encodes a ß1,4 mannosyltransferase that catalyzes the addition of the first of nine mannose moieties to form a dolichol-lipid linked oligosaccharide intermediate required for proper N-linked glycosylation. ALG1 mutations cause a rare autosomal recessive disorder termed ALG1-CDG. To date 13 mutations in 18 patients from 14 families have been described with varying degrees of clinical severity. We identified and characterized 39 previously unreported cases of ALG1-CDG from 32 families and add 26 new mutations. Pathogenicity of each mutation was confirmed based on its inability to rescue impaired growth or hypoglycosylation of a standard biomarker in an alg1-deficient yeast strain. Using this approach we could not establish a rank order comparison of biomarker glycosylation and patient phenotype, but we identified mutations with a lethal outcome in the first two years of life. The recently identified protein-linked xeno-tetrasaccharide biomarker, NeuAc-Gal-GlcNAc2 , was seen in all 27 patients tested. Our study triples the number of known patients and expands the molecular and clinical correlates of this disorder.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Mannosyltransferases/genetics , Mutation , Polysaccharides/metabolism , Biomarkers/metabolism , Congenital Disorders of Glycosylation/metabolism , Female , Genes, Lethal , Glycosylation , Humans , Male , Sequence Analysis, DNA , Survival Analysis
16.
Genet Med ; 18(11): 1090-1096, 2016 11.
Article in English | MEDLINE | ID: mdl-26938784

ABSTRACT

PURPOSE: To prospectively evaluate the diagnostic and clinical utility of singleton whole-exome sequencing (WES) as a first-tier test in infants with suspected monogenic disease. METHODS: Singleton WES was performed as a first-tier sequencing test in infants recruited from a single pediatric tertiary center. This occurred in parallel with standard investigations, including single- or multigene panel sequencing when clinically indicated. The diagnosis rate, clinical utility, and impact on management of singleton WES were evaluated. RESULTS: Of 80 enrolled infants, 46 received a molecular genetic diagnosis through singleton WES (57.5%) compared with 11 (13.75%) who underwent standard investigations in the same patient group. Clinical management changed following exome diagnosis in 15 of 46 diagnosed participants (32.6%). Twelve relatives received a genetic diagnosis following cascade testing, and 28 couples were identified as being at high risk of recurrence in future pregnancies. CONCLUSIONS: This prospective study provides strong evidence for increased diagnostic and clinical utility of singleton WES as a first-tier sequencing test for infants with a suspected monogenic disorder. Singleton WES outperformed standard care in terms of diagnosis rate and the benefits of a diagnosis, namely, impact on management of the child and clarification of reproductive risks for the extended family in a timely manner.Genet Med 18 11, 1090-1096.


Subject(s)
Genetic Diseases, Inborn/diagnosis , High-Throughput Nucleotide Sequencing/methods , Pathology, Molecular , Exome/genetics , Genetic Diseases, Inborn/genetics , Humans , Infant, Newborn
17.
N Engl J Med ; 373(11): 1010-20, 2015 Sep 10.
Article in English | MEDLINE | ID: mdl-26352813

ABSTRACT

BACKGROUND: Lysosomal acid lipase is an essential lipid-metabolizing enzyme that breaks down endocytosed lipid particles and regulates lipid metabolism. We conducted a phase 3 trial of enzyme-replacement therapy in children and adults with lysosomal acid lipase deficiency, an underappreciated cause of cirrhosis and severe dyslipidemia. METHODS: In this multicenter, randomized, double-blind, placebo-controlled study involving 66 patients, we evaluated the safety and effectiveness of enzyme-replacement therapy with sebelipase alfa (administered intravenously at a dose of 1 mg per kilogram of body weight every other week); the placebo-controlled phase of the study was 20 weeks long and was followed by open-label treatment for all patients. The primary end point was normalization of the alanine aminotransferase level. Secondary end points included additional disease-related efficacy assessments, safety, and side-effect profile. RESULTS: Substantial disease burden at baseline included a very high level of low-density lipoprotein cholesterol (≥190 mg per deciliter) in 38 of 66 patients (58%) and cirrhosis in 10 of 32 patients (31%) who underwent biopsy. A total of 65 of the 66 patients who underwent randomization completed the double-blind portion of the trial and continued with open-label treatment. At 20 weeks, the alanine aminotransferase level was normal in 11 of 36 patients (31%) in the sebelipase alfa group and in 2 of 30 (7%) in the placebo group (P=0.03), with mean changes from baseline of -58 U per liter versus -7 U per liter (P<0.001). With respect to prespecified key secondary efficacy end points, we observed improvements in lipid levels and reduction in hepatic fat content (P<0.001 for all comparisons, except P=0.04 for triglycerides). The number of patients with adverse events was similar in the two groups; most events were mild and were considered by the investigator to be unrelated to treatment. CONCLUSIONS: Sebelipase alfa therapy resulted in a reduction in multiple disease-related hepatic and lipid abnormalities in children and adults with lysosomal acid lipase deficiency. (Funded by Synageva BioPharma and others; ARISE ClinicalTrials.gov number, NCT01757184.).


Subject(s)
Sterol Esterase/therapeutic use , Wolman Disease/drug therapy , Adolescent , Adult , Aged , Alanine Transaminase/blood , Biopsy , Child , Child, Preschool , Cholesterol, HDL/blood , Cholesterol, LDL/blood , Double-Blind Method , Dyslipidemias/drug therapy , Dyslipidemias/genetics , Female , Humans , Liver/drug effects , Liver/pathology , Male , Middle Aged , Sterol Esterase/adverse effects , Sterol Esterase/pharmacology , Wolman Disease/blood , Young Adult , Wolman Disease
18.
Mol Genet Metab ; 115(4): 168-73, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26163321

ABSTRACT

3-Hydroxyisobutyryl-CoA hydrolase deficiency (HIBCHD) is a rare inborn error of the valine catabolic pathway associated with Leigh-like disease. We report a female patient who presented at the age of 5months with hypotonia, developmental delay and cerebral atrophy on MRI. Pyruvate dehydrogenase deficiency was initially suspected and decreased activity was shown in fibroblasts. Urine tandem mass spectrometry screening showed large increases in the cysteine conjugate of methacrylate previously described in HIBCHD. 3-hydroxyisobutyryl-CoA hydrolase activity in fibroblasts was below the limit of detection of the enzymatic assay and two novel HIBCH mutations were identified (c.[129dupA];[1033G>A]). Urine metabolite investigations also showed increases in 3-hydroxyisobutyryl carnitine, 2,3-dihydroxy-2-methylbutyrate and several metabolites indicating accumulation and subsequent metabolism of methacrylyl-CoA and acryloyl-CoA. The metabolites derived from acryloyl-CoA were also increased in patients with inborn errors of propionyl-CoA metabolism, indicating the involvement of a secondary propionyl-CoA pathway utilising 3-hydroxyisobutyryl-CoA hydrolase. With the exception of 3-hydroxyisobutyryl carnitine, the metabolite abnormalities were essentially the same as those observed in patients with ECHS1 mutations, a recently described disorder that also affects valine metabolism. Our findings demonstrate the benefits of urine tandem mass spectrometry screening for diagnosing HIBCH and ECHS1 defects and that propionate metabolism may play a role in their pathogenesis. These disorders should be considered during the differential diagnosis of Leigh like-diseases and hypotonia.


Subject(s)
Abnormalities, Multiple/urine , Amino Acid Metabolism, Inborn Errors/urine , Enoyl-CoA Hydratase/deficiency , Enoyl-CoA Hydratase/urine , Leigh Disease/diagnosis , Thiolester Hydrolases/deficiency , Abnormalities, Multiple/genetics , Amino Acid Metabolism, Inborn Errors/genetics , Child , Cysteine/analogs & derivatives , Cysteine/urine , Female , Fibroblasts/metabolism , Glutathione/metabolism , Humans , Infant , Leigh Disease/genetics , Mass Screening , Mutation , Prognosis , Thiolester Hydrolases/genetics , Thiolester Hydrolases/urine , Valine/metabolism
19.
Neuromuscul Disord ; 25(4): 321-32, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25617983

ABSTRACT

Emerging phenotypes in long-term survivors with Pompe disease on standard enzyme replacement therapy (ERT) (alglucosidase alfa 20 mg/kg/2 weeks) can include patients with worsening motor function. Whether higher doses of ERT improve skeletal function in these patients has not been systematically studied. This exploratory, randomized, open-label, 52-week study examined the safety and efficacy of 2 ERT regimens of alglucosidase alfa (20 mg/kg/week or 40 mg/kg/2 weeks) in 13 patients with Pompe disease and clinical decline or a lack of improvement on standard ERT: late-onset (n = 4), infantile-onset (n = 9). Cross-reactive immunologic material assay-negative patients were excluded. Eleven of 13 patients completed the study. Trends for improvement were seen in total gross motor function, but not mobility; however, 6 (late-onset, 2; infantile-onset, 4) of 11 patients (55%) who met the entry criteria of motor decline (late-onset, 4; infantile-onset, 7) showed improvement in motor and/or mobility skills. No between-regimen differences in efficacy emerged. Two case studies highlight the benefits of increased ERT dose in patients with Pompe disease experiencing clinical decline. Both alternative regimens were generally well tolerated. This study was limited by the small sample size, which is not uncommon for small clinical studies of rare diseases. Additionally, the study did not include direct assessment of muscle pathology, which may have identified potential causes of decreased response to ERT. Results were inconclusive but suggest that increased ERT dose may be beneficial in some patients with Pompe disease experiencing motor decline. Controlled studies are needed to clarify the benefits and risks of this strategy.


Subject(s)
Enzyme Replacement Therapy/methods , Glycogen Storage Disease Type II/drug therapy , Neuromuscular Agents/therapeutic use , alpha-Glucosidases/therapeutic use , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Cohort Studies , Enzyme Replacement Therapy/adverse effects , Female , Glycogen Storage Disease Type II/physiopathology , Humans , Immunoglobulin G/blood , Infant , Male , Middle Aged , Motor Skills/drug effects , Neuromuscular Agents/adverse effects , Random Allocation , Treatment Outcome , Young Adult , alpha-Glucosidases/adverse effects
SELECTION OF CITATIONS
SEARCH DETAIL
...