Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Inherit Metab Dis ; 47(2): 220-229, 2024 03.
Article in English | MEDLINE | ID: mdl-38375550

ABSTRACT

Carbamoyl phosphate synthetase 1 (CPS1) and ornithine transcarbamylase (OTC) deficiencies are rare urea cycle disorders, which can lead to life-threatening hyperammonemia. Liver transplantation (LT) provides a cure and offers an alternative to medical treatment and life-long dietary restrictions with permanent impending risk of hyperammonemia. Nevertheless, in most patients, metabolic aberrations persist after LT, especially low plasma citrulline levels, with questionable clinical impact. So far, little is known about these alterations and there is no consensus, whether l-citrulline substitution after LT improves patients' symptoms and outcomes. In this multicentre, retrospective, observational study of 24 patients who underwent LT for CPS1 (n = 11) or OTC (n = 13) deficiency, 25% did not receive l-citrulline or arginine substitution. Correlation analysis revealed no correlation between substitution dosage and citrulline levels (CPS1, p = 0.8 and OTC, p = 1). Arginine levels after liver transplantation were normal after LT independent of citrulline substitution. Native liver survival had no impact on mental impairment (p = 0.67). Regression analysis showed no correlation between l-citrulline substitution and failure to thrive (p = 0.611) or neurological outcome (p = 0.701). Peak ammonia had a significant effect on mental impairment (p = 0.017). Peak plasma ammonia levels correlate with mental impairment after LT in CPS1 and OTC deficiency. Growth and intellectual impairment after LT are not significantly associated with l-citrulline substitution.


Subject(s)
Hyperammonemia , Liver Transplantation , Ornithine Carbamoyltransferase Deficiency Disease , Humans , Ornithine Carbamoyltransferase Deficiency Disease/surgery , Hyperammonemia/drug therapy , Citrulline , Carbamyl Phosphate/metabolism , Carbamyl Phosphate/therapeutic use , Ammonia/metabolism , Retrospective Studies , Carbamoyl-Phosphate Synthase (Ammonia)/metabolism , Arginine/therapeutic use , Ornithine Carbamoyltransferase
2.
Mol Genet Metab ; 138(3): 107525, 2023 03.
Article in English | MEDLINE | ID: mdl-36796138

ABSTRACT

Glycogen storage disease type IV (GSD IV) is an ultra-rare autosomal recessive disorder caused by pathogenic variants in GBE1 which results in reduced or deficient glycogen branching enzyme activity. Consequently, glycogen synthesis is impaired and leads to accumulation of poorly branched glycogen known as polyglucosan. GSD IV is characterized by a remarkable degree of phenotypic heterogeneity with presentations in utero, during infancy, early childhood, adolescence, or middle to late adulthood. The clinical continuum encompasses hepatic, cardiac, muscular, and neurologic manifestations that range in severity. The adult-onset form of GSD IV, referred to as adult polyglucosan body disease (APBD), is a neurodegenerative disease characterized by neurogenic bladder, spastic paraparesis, and peripheral neuropathy. There are currently no consensus guidelines for the diagnosis and management of these patients, resulting in high rates of misdiagnosis, delayed diagnosis, and lack of standardized clinical care. To address this, a group of experts from the United States developed a set of recommendations for the diagnosis and management of all clinical phenotypes of GSD IV, including APBD, to support clinicians and caregivers who provide long-term care for individuals with GSD IV. The educational resource includes practical steps to confirm a GSD IV diagnosis and best practices for medical management, including (a) imaging of the liver, heart, skeletal muscle, brain, and spine, (b) functional and neuromusculoskeletal assessments, (c) laboratory investigations, (d) liver and heart transplantation, and (e) long-term follow-up care. Remaining knowledge gaps are detailed to emphasize areas for improvement and future research.


Subject(s)
Glycogen Storage Disease Type IV , Glycogen Storage Disease , Neurodegenerative Diseases , Child, Preschool , Humans , Glycogen Storage Disease Type IV/diagnosis , Glycogen Storage Disease Type IV/genetics , Glycogen Storage Disease Type IV/therapy , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/genetics , Glycogen Storage Disease/therapy , Glycogen
3.
Am J Hum Genet ; 105(2): 384-394, 2019 08 01.
Article in English | MEDLINE | ID: mdl-31256876

ABSTRACT

Proteins anchored to the cell surface via glycosylphosphatidylinositol (GPI) play various key roles in the human body, particularly in development and neurogenesis. As such, many developmental disorders are caused by mutations in genes involved in the GPI biosynthesis and remodeling pathway. We describe ten unrelated families with bi-allelic mutations in PIGB, a gene that encodes phosphatidylinositol glycan class B, which transfers the third mannose to the GPI. Ten different PIGB variants were found in these individuals. Flow cytometric analysis of blood cells and fibroblasts from the affected individuals showed decreased cell surface presence of GPI-anchored proteins. Most of the affected individuals have global developmental and/or intellectual delay, all had seizures, two had polymicrogyria, and four had a peripheral neuropathy. Eight children passed away before four years old. Two of them had a clinical diagnosis of DOORS syndrome (deafness, onychodystrophy, osteodystrophy, mental retardation, and seizures), a condition that includes sensorineural deafness, shortened terminal phalanges with small finger and toenails, intellectual disability, and seizures; this condition overlaps with the severe phenotypes associated with inherited GPI deficiency. Most individuals tested showed elevated alkaline phosphatase, which is a characteristic of the inherited GPI deficiency but not DOORS syndrome. It is notable that two severely affected individuals showed 2-oxoglutaric aciduria, which can be seen in DOORS syndrome, suggesting that severe cases of inherited GPI deficiency and DOORS syndrome might share some molecular pathway disruptions.


Subject(s)
Craniofacial Abnormalities/etiology , Glycosylphosphatidylinositols/biosynthesis , Glycosylphosphatidylinositols/deficiency , Hand Deformities, Congenital/etiology , Hearing Loss, Sensorineural/etiology , Intellectual Disability/etiology , Mannosyltransferases/genetics , Metabolic Diseases/etiology , Mutation , Nails, Malformed/etiology , Peripheral Nervous System Diseases/etiology , Seizures/pathology , Adult , Child , Child, Preschool , Craniofacial Abnormalities/pathology , Female , Glycosylphosphatidylinositols/genetics , Hand Deformities, Congenital/pathology , Hearing Loss, Sensorineural/pathology , Humans , Infant , Infant, Newborn , Intellectual Disability/pathology , Male , Metabolic Diseases/pathology , Nails, Malformed/pathology , Pedigree , Peripheral Nervous System Diseases/pathology , Seizures/genetics , Severity of Illness Index , Young Adult
4.
Am J Hum Genet ; 99(3): 720-727, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545676

ABSTRACT

SON is a key component of the spliceosomal complex and a critical mediator of constitutive and alternative splicing. Additionally, SON has been shown to influence cell-cycle progression, genomic integrity, and maintenance of pluripotency in stem cell populations. The clear functional relevance of SON in coordinating essential cellular processes and its presence in diverse human tissues suggests that intact SON might be crucial for normal growth and development. However, the phenotypic effects of deleterious germline variants in SON have not been clearly defined. Herein, we describe seven unrelated individuals with de novo variants in SON and propose that deleterious variants in SON are associated with a severe multisystem disorder characterized by developmental delay, persistent feeding difficulties, and congenital malformations, including brain anomalies.


Subject(s)
Congenital Abnormalities/genetics , DNA-Binding Proteins/genetics , Developmental Disabilities/genetics , Failure to Thrive/genetics , Intellectual Disability/genetics , Minor Histocompatibility Antigens/genetics , Sequence Deletion/genetics , Adolescent , Brain/abnormalities , Child , Child, Preschool , DNA-Binding Proteins/chemistry , Exome/genetics , Female , Humans , Male , Minor Histocompatibility Antigens/chemistry , Pedigree , Young Adult
5.
Genet Med ; 21(4): 772-789, 2019 04.
Article in English | MEDLINE | ID: mdl-30659246

ABSTRACT

PURPOSE: Glycogen storage disease (GSD) types VI and IX are rare diseases of variable clinical severity affecting primarily the liver. GSD VI is caused by deficient activity of hepatic glycogen phosphorylase, an enzyme encoded by the PYGL gene. GSD IX is caused by deficient activity of phosphorylase kinase (PhK), the enzyme subunits of which are encoded by various genes: ɑ (PHKA1, PHKA2), ß (PHKB), É£ (PHKG1, PHKG2), and δ (CALM1, CALM2, CALM3). Glycogen storage disease types VI and IX have a wide spectrum of clinical manifestations and often cannot be distinguished from each other, or from other liver GSDs, on clinical presentation alone. Individuals with GSDs VI and IX can present with hepatomegaly with elevated serum transaminases, ketotic hypoglycemia, hyperlipidemia, and poor growth. This guideline for the management of GSDs VI and IX was developed as an educational resource for health-care providers to facilitate prompt and accurate diagnosis and appropriate management of patients. METHODS: A national group of experts in various aspects of GSDs VI and IX met to review the limited evidence base from the scientific literature and provided their expert opinions. Consensus was developed in each area of diagnosis, treatment, and management. Evidence bases for these rare disorders are largely based on expert opinion, particularly when targeted therapeutics that have to clear the US Food and Drug Administration (FDA) remain unavailable. RESULTS: This management guideline specifically addresses evaluation and diagnosis across multiple organ systems involved in GSDs VI and IX. Conditions to consider in a differential diagnosis stemming from presenting features and diagnostic algorithms are discussed. Aspects of diagnostic evaluation and nutritional and medical management, including care coordination, genetic counseling, and prenatal diagnosis are addressed. CONCLUSION: A guideline that will facilitate the accurate diagnosis and optimal management of patients with GSDs VI and IX was developed. This guideline will help health-care providers recognize patients with GSDs VI and IX, expedite diagnosis, and minimize adverse sequelae from delayed diagnosis and inappropriate management. It will also help identify gaps in scientific knowledge that exist today and suggest future studies.


Subject(s)
Genomics , Glycogen Storage Disease/genetics , Hypoglycemia/genetics , Phosphorylase Kinase/genetics , Disease Management , Genetics, Medical/trends , Glycogen/genetics , Glycogen/metabolism , Glycogen Storage Disease/diagnosis , Glycogen Storage Disease/epidemiology , Glycogen Storage Disease/therapy , Guidelines as Topic , Humans , Hypoglycemia/metabolism , Hypoglycemia/therapy , Liver/metabolism , Liver/pathology , Mutation , Phosphorylase Kinase/chemistry , United States/epidemiology
6.
J Inherit Metab Dis ; 41(2): 169-180, 2018 03.
Article in English | MEDLINE | ID: mdl-29238895

ABSTRACT

Combined D-2- and L-2-hydroxyglutaric aciduria (D/L-2-HGA) is a devastating neurometabolic disorder, usually lethal in the first years of life. Autosomal recessive mutations in the SLC25A1 gene, which encodes the mitochondrial citrate carrier (CIC), were previously detected in patients affected with combined D/L-2-HGA. We showed that transfection of deficient fibroblasts with wild-type SLC25A1 restored citrate efflux and decreased intracellular 2-hydroxyglutarate levels, confirming that deficient CIC is the cause of D/L-2-HGA. We developed and implemented a functional assay and applied it to all 17 missense variants detected in a total of 26 CIC-deficient patients, including eight novel cases, showing reduced activities of varying degrees. In addition, we analyzed the importance of residues affected by these missense variants using our existing scoring system. This allowed not only a clinical and biochemical overview of the D/L-2-HGA patients but also phenotype-genotype correlation studies.


Subject(s)
Anion Transport Proteins/metabolism , Brain Diseases, Metabolic, Inborn/metabolism , Citric Acid/metabolism , Glutarates/metabolism , Mitochondrial Proteins/metabolism , Anion Transport Proteins/chemistry , Anion Transport Proteins/genetics , Biological Assay/methods , Brain Diseases, Metabolic, Inborn/genetics , Cells, Cultured , Child, Preschool , DNA Mutational Analysis , Female , Fibroblasts , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Male , Mitochondrial Proteins/chemistry , Mitochondrial Proteins/genetics , Models, Molecular , Mutation, Missense , Organic Anion Transporters , Phenotype , Protein Conformation , Structure-Activity Relationship
7.
Cleft Palate Craniofac J ; 55(5): 773-777, 2018 05.
Article in English | MEDLINE | ID: mdl-29489401

ABSTRACT

Pierre Robin Sequence (PRS) can be associated with skeletal dysplasias, presenting with craniocervical instability and devastating spinal injury if unrecognized. The authors present the case of an infant with PRS and a type II collagenopathy who underwent multiple airway-securing procedures requiring spinal manipulation before craniocervical instability was identified. This resulted in severe cervical cord compression due to odontoid fracture and occipitoatlantoaxial instability. This case highlights the importance of early cervical spine imaging and cautious manipulation in infants with PRS and suspected skeletal dysplasia.


Subject(s)
Airway Obstruction/surgery , Atlanto-Axial Joint/injuries , Joint Instability/etiology , Odontoid Process/injuries , Osteochondrodysplasias/etiology , Patient Positioning/adverse effects , Pierre Robin Syndrome/complications , Pierre Robin Syndrome/surgery , Plastic Surgery Procedures/methods , Airway Obstruction/diagnostic imaging , Atlanto-Axial Joint/diagnostic imaging , Humans , Infant, Newborn , Joint Instability/diagnostic imaging , Joint Instability/surgery , Magnetic Resonance Imaging , Male , Odontoid Process/diagnostic imaging , Odontoid Process/surgery , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/surgery , Pierre Robin Syndrome/diagnostic imaging , Plastic Surgery Procedures/adverse effects , Spinal Cord Compression/diagnostic imaging , Spinal Cord Compression/etiology , Spinal Cord Compression/surgery , Tomography, X-Ray Computed , Treatment Outcome
8.
Am J Hum Genet ; 95(5): 579-83, 2014 Nov 06.
Article in English | MEDLINE | ID: mdl-25439098

ABSTRACT

5q31.3 microdeletion syndrome is characterized by neonatal hypotonia, encephalopathy with or without epilepsy, and severe developmental delay, and the minimal critical deletion interval harbors three genes. We describe 11 individuals with clinical features of 5q31.3 microdeletion syndrome and de novo mutations in PURA, encoding transcriptional activator protein Pur-α, within the critical region. These data implicate causative PURA mutations responsible for the severe neurological phenotypes observed in this syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Chromosome Deletion , Chromosomes, Human, Pair 5/genetics , DNA-Binding Proteins/genetics , Muscle Hypotonia/genetics , Seizures/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Base Sequence , Caenorhabditis elegans/genetics , Chromosome Mapping , Humans , Molecular Sequence Data , Mutation/genetics , Sequence Analysis, DNA , Syndrome
9.
J Inherit Metab Dis ; 40(6): 831-843, 2017 11.
Article in English | MEDLINE | ID: mdl-28871440

ABSTRACT

BACKGROUND: Observational reports suggest that supplementation that increases citric acid cycle intermediates via anaplerosis may have therapeutic advantages over traditional medium-chain triglyceride (MCT) treatment of long-chain fatty acid oxidation disorders (LC-FAODs) but controlled trials have not been reported. The goal of our study was to compare the effects of triheptanoin (C7), an anaplerotic seven-carbon fatty acid triglyceride, to trioctanoin (C8), an eight-carbon fatty acid triglyceride, in patients with LC-FAODs. METHODS: A double blinded, randomized controlled trial of 32 subjects with LC-FAODs (carnitine palmitoyltransferase-2, very long-chain acylCoA dehydrogenase, trifunctional protein or long-chain 3-hydroxy acylCoA dehydrogenase deficiencies) who were randomly assigned a diet containing 20% of their total daily energy from either C7 or C8 for 4 months was conducted. Primary outcomes included changes in total energy expenditure (TEE), cardiac function by echocardiogram, exercise tolerance, and phosphocreatine recovery following acute exercise. Secondary outcomes included body composition, blood biomarkers, and adverse events, including incidence of rhabdomyolysis. RESULTS: Patients in the C7 group increased left ventricular (LV) ejection fraction by 7.4% (p = 0.046) while experiencing a 20% (p = 0.041) decrease in LV wall mass on their resting echocardiogram. They also required a lower heart rate for the same amount of work during a moderate-intensity exercise stress test when compared to patients taking C8. There was no difference in TEE, phosphocreatine recovery, body composition, incidence of rhabdomyolysis, or any secondary outcome measures between the groups. CONCLUSIONS: C7 improved LV ejection fraction and reduced LV mass at rest, as well as lowering heart rate during exercise among patients with LC-FAODs. CLINICAL TRIAL REGISTRATION: Clinicaltrials.gov NCT01379625.


Subject(s)
Caprylates/therapeutic use , Cardiomyopathies/drug therapy , Fatty Acids/metabolism , Lipid Metabolism, Inborn Errors/drug therapy , Mitochondrial Myopathies/drug therapy , Mitochondrial Trifunctional Protein/deficiency , Nervous System Diseases/drug therapy , Rhabdomyolysis/drug therapy , Triglycerides/therapeutic use , Acyl-CoA Dehydrogenase, Long-Chain/metabolism , Adolescent , Adult , Cardiomyopathies/metabolism , Carnitine/metabolism , Child , Dietary Fats/metabolism , Double-Blind Method , Exercise/physiology , Female , Humans , Lipid Metabolism, Inborn Errors/metabolism , Male , Middle Aged , Mitochondrial Myopathies/metabolism , Mitochondrial Trifunctional Protein/metabolism , Nervous System Diseases/metabolism , Oxidation-Reduction , Rhabdomyolysis/metabolism , Young Adult
10.
Mol Genet Metab ; 109(2): 215-7, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23583224

ABSTRACT

Guanidinoacetate methyltransferase (GAMT) deficiency is a good candidate disorder for newborn screening because early treatment appears to improve outcomes. We report elevation of guanidinoacetate in archived newborn dried blood spots for 3 cases (2 families) of GAMT deficiency compared with an unaffected carrier and controls. We also report a new case of a patient treated from birth with normal developmental outcome at the age of 42 months.


Subject(s)
Glycine/analogs & derivatives , Guanidinoacetate N-Methyltransferase/deficiency , Language Development Disorders/therapy , Movement Disorders/congenital , Case-Control Studies , Child, Preschool , Creatine/therapeutic use , Dried Blood Spot Testing , Early Diagnosis , Female , Glycine/blood , Guanidinoacetate N-Methyltransferase/blood , Humans , Infant , Infant, Newborn , Language Development Disorders/blood , Language Development Disorders/diagnosis , Male , Movement Disorders/blood , Movement Disorders/diagnosis , Movement Disorders/therapy , Ornithine/therapeutic use , Treatment Outcome
11.
Sci Transl Med ; 15(692): eadf4086, 2023 04 19.
Article in English | MEDLINE | ID: mdl-37075130

ABSTRACT

Glutaric aciduria type I (GA-1) is an inborn error of metabolism with a severe neurological phenotype caused by the deficiency of glutaryl-coenzyme A dehydrogenase (GCDH), the last enzyme of lysine catabolism. Current literature suggests that toxic catabolites in the brain are produced locally and do not cross the blood-brain barrier. In a series of experiments using knockout mice of the lysine catabolic pathway and liver cell transplantation, we uncovered that toxic GA-1 catabolites in the brain originated from the liver. Moreover, the characteristic brain and lethal phenotype of the GA-1 mouse model was rescued by two different liver-directed gene therapy approaches: Using an adeno-associated virus, we replaced the defective Gcdh gene or we prevented flux through the lysine degradation pathway by CRISPR deletion of the aminoadipate-semialdehyde synthase (Aass) gene. Our findings question the current pathophysiological understanding of GA-1 and reveal a targeted therapy for this devastating disorder.


Subject(s)
Glutaryl-CoA Dehydrogenase , Lysine , Animals , Mice , Glutaryl-CoA Dehydrogenase/genetics , Glutaryl-CoA Dehydrogenase/metabolism , Lysine/metabolism , Mice, Knockout , Liver/metabolism
12.
Mol Genet Metab Rep ; 31: 100856, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35782603

ABSTRACT

Introduction: Biotinidase synthesis is needed to recycle biotin for essential metabolic reactions. Biotinidase activity is lower than normal levels in advanced liver disease but is higher in hepatic glycogen storage disorders (GSDs), however the cause of this association remains unclear. Methods: In this study, biotinidase activity was measured in plasma samples from 45 individuals with hepatic GSDs; GSDI (a, b; n = 25) and GSD III (a, b; n = 20), complemented by a chart review to associate biotinidase activity levels with clinical laboratory and imaging findings known to be implicated in these GSDs. Results: Our findings showed variation in biotinidase activity levels among subjects with GSD I and III; biotinidase activity correlated positively with hypertriglyceridemia in subjects with GSD I (r = 0.47, P = 0.036) and GSD III (r = 0.58, P = 0.014), and correlated negatively with age (r = -0.50, P = 0.03) in patients with GSD III. Additionally, biotinidase activity was reduced, albeit within the normal range in subjects with evidence of fibrosis/cirrhosis, as compared to subjects with hepatomegaly with or without steatosis (P = 0.002). Discussions: These findings suggest that abnormal lipid metabolism in GSD I and III and progressive liver disease in GSD III may influence biotinidase activity levels. We suggest that a prospective, multi-center, longitudinal study designed to assess the significance of monitoring biotinidase activity in a larger cohort with hepatic GSDs is warranted to confirm this observation. Take-home message: Altered lipid metabolism and advancing liver fibrosis/cirrhosis may influence biotinidase activity levels in patients with hepatic glycogen storage disease. Thus, longitudinal monitoring of biotinidase activity, when combined with clinical and other biochemical findings may be informative.

13.
Mol Genet Metab ; 102(2): 214-5, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21130013

ABSTRACT

A male child with X-linked pyruvate dehydrogenase deficiency presented with severe neonatal lactic acidosis. Poor compliance following initiation of the ketogenic diet justified modification to a less restrictive form which improved compliance. One year after starting the modified diet, he remained clinically stable, showing developmental progress.


Subject(s)
Diet, Ketogenic , Pyruvate Dehydrogenase Complex Deficiency Disease/diet therapy , Child, Preschool , Diet, Ketogenic/standards , Follow-Up Studies , Humans , Hydroxybutyrates/blood , Lactic Acid/blood , Male , Patient Compliance , Treatment Outcome
14.
Mol Genet Metab ; 103(4): 362-6, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21605996

ABSTRACT

PURPOSE: Cerebro-vascular arteriopathy has been reported in late-onset Pompe disease (LOPD). Evidence of increased aortic stiffness in some patients and smooth muscle involvement in LOPD raises the possibility of aortic involvement. Our aim was to determine if aortic arteriopathy may be a complication of LOPD. METHODS: One patient with LOPD was diagnosed with aortic dilatation at Duke Metabolic clinic, 4 others were diagnosed at University of Mainz, Germany, where chest X-ray and echocardiography are routinely done for patients. Other causes of aortic vascular disease were assessed. RESULTS: We report evidence of dilated arteriopathy involving primarily the ascending thoracic aorta in 5 females with late-onset Pompe disease. One patient had a bicuspid aortic valve and developed dissection. Another patient with juvenile onset disease had both thoracic and basilar artery aneurysms. CONCLUSIONS: Aneurysmal dilatation of the thoracic aorta is an underreported vascular complication of LOPD, probably due to the same pathological process that occurs in the brain. Chest X-ray together with echocardiography should be incorporated as initial screening tools for aortic aneurysms in patients with LOPD. When ectasia is suspected, or the ascending aorta is not visualized, contrast- mediated thoracic CT or MRA may be necessary. Large-scale studies are warranted to determine the prevalence and extent of aortic vascular involvement.


Subject(s)
Aorta, Thoracic/pathology , Aortic Diseases/pathology , Glycogen Storage Disease Type II/pathology , Adult , Child, Preschool , Dilatation, Pathologic , Female , Humans , Middle Aged , Phenotype
15.
Mol Genet Metab ; 104(1-2): 118-22, 2011.
Article in English | MEDLINE | ID: mdl-21802969

ABSTRACT

PURPOSE: Pompe disease (PD) is a progressive metabolic myopathy for which the only available treatment is alglucosidase alfa (Myozyme®). Enzyme replacement therapy (ERT) has improved ventilator-free survival, and cardiac and motor functions in patients with infantile PD. However, for an adequate response to occur, a large amount of enzymes must be infused. In some patients, this may be problematic due to infusion-associated reactions (IARs) occurring in approximately 50% of patients receiving alglucosidase alfa infusions. Whilst the majority of these reactions are mild, life threatening hypersensitivity reactions may occur in some patients. In these patients desensitization is indicated to enable continued ERT safely. Infants and young children with PD and significant infusion reactions pose unique management challenges because of their young age, limited communication skills, variable presentation and underlying cardiomyopathy. METHODS/SUBJECTS: In 2 patients with PD who experienced significant ERT-related reactions: an infant (IgE positive) and a young child (IgE negative), we implemented a desensitization protocol, that started by administering a reduced dose of alglucosidase alfa (10 mg/kg weekly) instead of the standard (20 mg/kg bi-weekly) using serial micro-dilutions that were individually prepared and delivered in a highly regulated manner based on patients' clinical manifestations and tolerance. RESULTS: Successful desensitization was achieved in both patients, allowing them to eventually continue to receive the full dose of ERT safely. CONCLUSION: Therapeutic demands in infants and young children with PD need to be tailored according to the patient presentation, and underlying cardiac and fluid-volume status. Desensitization allowed both patients to continue alglucosidase alfa treatment at the recommended dose without prolonged interruption of therapy, or further reactions.


Subject(s)
Desensitization, Immunologic , Enzyme Replacement Therapy/adverse effects , Glycogen Storage Disease Type II/drug therapy , Glycogen Storage Disease Type II/immunology , Precision Medicine , alpha-Glucosidases/adverse effects , alpha-Glucosidases/therapeutic use , Child , Humans , Immunoglobulin E/immunology , Infant , Infusions, Intravenous , Male , Recurrence
16.
Mol Genet Metab Rep ; 29: 100821, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34820282

ABSTRACT

INTRODUCTION: A deficiency of glycogen debrancher enzyme in patients with glycogen storage disease type III (GSD III) manifests with hepatic, cardiac, and muscle involvement in the most common subtype (type a), or with only hepatic involvement in patients with GSD IIIb. OBJECTIVE AND METHODS: To describe longitudinal biochemical, radiological, muscle strength and ambulation, liver histopathological findings, and clinical outcomes in adults (≥18 years) with glycogen storage disease type III, by a retrospective review of medical records. RESULTS: Twenty-one adults with GSD IIIa (14 F & 7 M) and four with GSD IIIb (1 F & 3 M) were included in this natural history study. At the most recent visit, the median (range) age and follow-up time were 36 (19-68) and 16 years (0-41), respectively. For the entire cohort: 40% had documented hypoglycemic episodes in adulthood; hepatomegaly and cirrhosis were the most common radiological findings; and 28% developed decompensated liver disease and portal hypertension, the latter being more prevalent in older patients. In the GSD IIIa group, muscle weakness was a major feature, noted in 89% of the GSD IIIa cohort, a third of whom depended on a wheelchair or an assistive walking device. Older individuals tended to show more severe muscle weakness and mobility limitations, compared with younger adults. Asymptomatic left ventricular hypertrophy (LVH) was the most common cardiac manifestation, present in 43%. Symptomatic cardiomyopathy and reduced ejection fraction was evident in 10%. Finally, a urinary biomarker of glycogen storage (Glc4) was significantly associated with AST, ALT and CK. CONCLUSION: GSD III is a multisystem disorder in which a multidisciplinary approach with regular clinical, biochemical, radiological and functional (physical therapy assessment) follow-up is required. Despite dietary modification, hepatic and myopathic disease progression is evident in adults, with muscle weakness as the major cause of morbidity. Consequently, definitive therapies that address the underlying cause of the disease to correct both liver and muscle are needed.

17.
Genet Med ; 12(7): 446-63, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20631546

ABSTRACT

PURPOSE: Glycogen storage disease type III is a rare disease of variable clinical severity affecting primarily the liver, heart, and skeletal muscle. It is caused by deficient activity of glycogen debranching enzyme, which is a key enzyme in glycogen degradation. Glycogen storage disease type III manifests a wide clinical spectrum. Individuals with glycogen storage disease type III present with hepatomegaly, hypoglycemia, hyperlipidemia, and growth retardation. Those with type IIIa have symptoms related to liver disease and progressive muscle (cardiac and skeletal) involvement that varies in age of onset, rate of disease progression, and severity. Those with type IIIb primarily have symptoms related to liver disease. This guideline for the management of glycogen storage disease type III was developed as an educational resource for health care providers to facilitate prompt and accurate diagnosis and appropriate management of patients. METHODS: An international group of experts in various aspects of glycogen storage disease type III met to review the evidence base from the scientific literature and provided their expert opinions. Consensus was developed in each area of diagnosis, treatment, and management. RESULTS: This management guideline specifically addresses evaluation and diagnosis across multiple organ systems (cardiovascular, gastrointestinal/nutrition, hepatic, musculoskeletal, and neuromuscular) involved in glycogen storage disease type III. Conditions to consider in a differential diagnosis stemming from presenting features and diagnostic algorithms are discussed. Aspects of diagnostic evaluation and nutritional and medical management, including care coordination, genetic counseling, hepatic transplantation, and prenatal diagnosis, are addressed. CONCLUSIONS: A guideline that will facilitate the accurate diagnosis and appropriate management of individuals with glycogen storage disease type III was developed. This guideline will help health care providers recognize patients with all forms of glycogen storage disease type III, expedite diagnosis, and minimize stress and negative sequelae from delayed diagnosis and inappropriate management. It will also help identify gaps in scientific knowledge that exist today and suggest future studies.


Subject(s)
Glycogen Storage Disease Type III/diagnosis , Glycogen Storage Disease Type III/therapy , Liver/pathology , Muscle, Skeletal/pathology , Humans , Liver/metabolism , Muscle, Skeletal/metabolism , Prognosis
18.
Am J Med Genet A ; 152A(1): 169-74, 2010 Jan.
Article in English | MEDLINE | ID: mdl-20014132

ABSTRACT

Cleidocranial dysplasia (CCD) is a rare autosomal dominant skeletal dysplasia due to mutations causing haploinsufficiency of RUNX2, an osteoblast transcription factor specific for bone and cartilage. The classic form of CCD is characterized by delayed closure of the fontanels, hypoplastic or aplastic clavicles and dental anomalies. Clinical reports suggest that a subset of patients with CCD have skeletal changes which mimic hypophosphatasia (HPP). Mutations in RUNX2 are detected in approximately 65% of cases of CCD, and microdeletions occur in 13%. We present clinical and radiological features in a 6-year-old child with severe CCD manifested by absence of the clavicles marked calvarial hypomineralization, osteoporosis and progressive kyphoscoliosis. HPP features included Bowdler spurs, severe osteopenia, and low alkaline phosphatase. Following negative mutation analysis of RUNX2, comparative genomic hybridization (CGH) microarray was performed. The result revealed a microdeletion in RUNX2, disrupting the C-terminal part of the gene.


Subject(s)
Cleidocranial Dysplasia/genetics , Core Binding Factor Alpha 1 Subunit/genetics , Hypophosphatasia/genetics , Sequence Deletion , Child , Humans , Male
19.
Pediatr Clin North Am ; 65(2): 317-335, 2018 04.
Article in English | MEDLINE | ID: mdl-29502916

ABSTRACT

Fatty acid oxidation disorders (FAODs) and carnitine shuttling defects are inborn errors of energy metabolism with associated mortality and morbidity due to cardiomyopathy, exercise intolerance, rhabdomyolysis, and liver disease with physiologic stress. Hypoglycemia is characteristically hypoketotic. Lactic acidemia and hyperammonemia may occur during decompensation. Recurrent rhabdomyolysis is debilitating. Expanded newborn screening can detect most of these disorders, allowing early, presymptomatic treatment. Treatment includes avoiding fasting and sustained extraneous exercise and providing high-calorie hydration during illness to prevent lipolysis, and medium-chain triglyceride oil supplementation in long-chain FAODs. Carnitine supplementation may be helpful. However, conventional treatment does not prevent all symptoms.


Subject(s)
Carnitine/metabolism , Fatty Acids/metabolism , Metabolism, Inborn Errors/diagnosis , Muscular Diseases/etiology , Neonatal Screening/methods , Humans , Infant , Infant, Newborn , Metabolism, Inborn Errors/complications , Metabolism, Inborn Errors/therapy , Muscular Diseases/therapy , Oxidation-Reduction
20.
Ann Transl Med ; 6(24): 474, 2018 Dec.
Article in English | MEDLINE | ID: mdl-30740405

ABSTRACT

Glucose is the main energy fuel for the human brain. Maintenance of glucose homeostasis is therefore, crucial to meet cellular energy demands in both - normal physiological states and during stress or increased demands. Glucose is stored as glycogen primarily in the liver and skeletal muscle with a small amount stored in the brain. Liver glycogen primarily maintains blood glucose levels, while skeletal muscle glycogen is utilized during high-intensity exertion, and brain glycogen is an emergency cerebral energy source. Glycogen and glucose transform into one another through glycogen synthesis and degradation pathways. Thus, enzymatic defects along these pathways are associated with altered glucose metabolism and breakdown leading to hypoglycemia ± hepatomegaly and or liver disease in hepatic forms of glycogen storage disorder (GSD) and skeletal ± cardiac myopathy, depending on the site of the enzyme defects. Overall, defects in glycogen metabolism mainly present as GSDs and are a heterogenous group of inborn errors of carbohydrate metabolism. In this article we review the genetics, epidemiology, clinical and metabolic findings of various types of GSD, and glycolysis defects emphasizing current treatment and implications for future directions.

SELECTION OF CITATIONS
SEARCH DETAIL