Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
1.
Mol Brain ; 14(1): 91, 2021 06 13.
Article in English | MEDLINE | ID: mdl-34120625

ABSTRACT

N-glycanase 1 (NGLY1) deficiency is a rare inherited disorder characterized by developmental delay, hypolacrima or alacrima, seizure, intellectual disability, motor deficits, and other neurological symptoms. The underlying mechanisms of the NGLY1 phenotype are poorly understood, and no effective therapy is currently available. Similar to human patients, the rat model of NGLY1 deficiency, Ngly1-/-, shows developmental delay, movement disorder, somatosensory impairment, scoliosis, and learning disability. Here we show that single intracerebroventricular administration of AAV9 expressing human NGLY1 cDNA (AAV9-hNGLY1) to Ngly1-/- rats during the weaning period restored NGLY1 expression in the brain and spinal cord, concomitant with increased enzymatic activity of NGLY1 in the brain. hNGLY1 protein expressed by AAV9 was found predominantly in mature neurons, but not in glial cells, of Ngly1-/- rats. Strikingly, intracerebroventricular administration of AAV9-hNGLY1 normalized the motor phenotypes of Ngly1-/- rats assessed by the rota-rod test and gait analysis. The reversibility of motor deficits in Ngly1-/- rats by central nervous system (CNS)-restricted gene delivery suggests that the CNS is the primary therapeutic target organs for NGLY1 deficiency, and that the Ngly1-/- rat model may be useful for evaluating therapeutic treatments in pre-clinical studies.


Subject(s)
Congenital Disorders of Glycosylation/physiopathology , Motor Activity/physiology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Acetylglucosamine/analogs & derivatives , Animals , Congenital Disorders of Glycosylation/enzymology , Disease Models, Animal , Genetic Therapy , Genetic Vectors/administration & dosage , Gliosis/complications , Gliosis/pathology , Humans , Inflammation/pathology , Injections, Intraventricular , Neurons/pathology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Rats , Rats, Transgenic , Transgenes
2.
Clin Genet ; 100(3): 318-323, 2021 09.
Article in English | MEDLINE | ID: mdl-33960418

ABSTRACT

Congenital disorders of glycosylation (CDG) are a heterogeneous group of genetic defects in glycoprotein and glycolipid glycan synthesis and attachment. A CDG subgroup are defects in the conserved oligomeric Golgi complex encoded by eight genes, COG1-COG8. Pathogenic variants in all genes except the COG3 gene have been reported. COG1-CDG has been reported in five patients. We report a male with neonatal seizures, dysmorphism, hepatitis and a type 2 serum transferrin isoelectrofocusing. Exome sequencing identified a homozygous COG1 variant (NM_018714.3: c.2665dup: p.[Arg889Profs*12]), which has been reported previously in one patient. We review the reported patients.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/physiopathology , Adaptor Proteins, Vesicular Transport/genetics , Congenital Disorders of Glycosylation/pathology , Frameshift Mutation , Glycosylation , Humans , Infant, Newborn , Male , Exome Sequencing
3.
J Inherit Metab Dis ; 44(4): 1001-1012, 2021 07.
Article in English | MEDLINE | ID: mdl-33734437

ABSTRACT

Pathogenic variants in ALG13 (ALG13 UDP-N-acetylglucosaminyltransferase subunit) cause an X-linked congenital disorder of glycosylation (ALG13-CDG) where individuals have variable clinical phenotypes that include developmental delay, intellectual disability, infantile spasms, and epileptic encephalopathy. Girls with a recurrent de novo c.3013C>T; p.(Asn107Ser) variant have normal transferrin glycosylation. Using a highly sensitive, semi-quantitative flow injection-electrospray ionization-quadrupole time-of-flight mass spectrometry (ESI-QTOF/MS) N-glycan assay, we report subtle abnormalities in N-glycans that normally account for <0.3% of the total plasma glycans that may increase up to 0.5% in females with the p.(Asn107Ser) variant. Among our 11 unrelated ALG13-CDG individuals, one male had abnormal serum transferrin glycosylation. We describe seven previously unreported subjects including three novel variants in ALG13 and report a milder neurodevelopmental course. We also summarize the molecular, biochemical, and clinical data for the 53 previously reported ALG13-CDG individuals. We provide evidence that ALG13 pathogenic variants may mildly alter N-linked protein glycosylation in both female and male subjects, but the underlying mechanism remains unclear.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Intellectual Disability/physiopathology , N-Acetylglucosaminyltransferases/genetics , Congenital Disorders of Glycosylation/physiopathology , Female , Genetic Variation , Glycosylation , Humans , Intellectual Disability/genetics , Male , Phenotype , Transferrin/metabolism
4.
J Inherit Metab Dis ; 44(4): 987-1000, 2021 07.
Article in English | MEDLINE | ID: mdl-33583022

ABSTRACT

Congenital disorders of glycosylation (CDGs) are a continuously expanding group of monogenic disorders of glycoprotein and glycolipid biosynthesis that cause multisystem diseases. Individuals with ALG3-CDG frequently exhibit severe neurological involvement (epilepsy, microcephaly, and hypotonia), ocular anomalies, dysmorphic features, skeletal anomalies, and feeding difficulties. We present 10 unreported individuals diagnosed with ALG3-CDG based on molecular and biochemical testing with 11 novel variants in ALG3, bringing the total to 40 reported individuals. In addition to the typical multisystem disease seen in ALG3-CDG, we expand the symptomatology of ALG3-CDG to now include endocrine abnormalities, neural tube defects, mild aortic root dilatation, immunodeficiency, and renal anomalies. N-glycan analyses of these individuals showed combined deficiencies of hybrid glycans and glycan extension beyond Man5 GlcNAc2 consistent with their truncated lipid-linked precursor oligosaccharides. This spectrum of N-glycan changes is unique to ALG3-CDG. These expanded features of ALG3-CDG facilitate diagnosis and suggest that optimal management should include baseline endocrine, renal, cardiac, and immunological evaluation at the time of diagnosis and with ongoing monitoring.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Mannosyltransferases/genetics , Adolescent , Adult , Child, Preschool , Congenital Disorders of Glycosylation/pathology , Congenital Disorders of Glycosylation/physiopathology , Female , Genotype , Humans , Infant , Infant, Newborn , Male , Phenotype , Young Adult
5.
Am J Med Genet A ; 185(4): 1187-1194, 2021 04.
Article in English | MEDLINE | ID: mdl-33394555

ABSTRACT

Congenital disorders of glycosylation (CDG) are an expanding group of metabolic disorders that result from abnormal protein glycosylation. A special subgroup of CDG type II comprises defects in the Conserved Oligomeric Golgi Complex (COG). In order to further delineate the genotypic and phenotypic spectrum of COG complex defect, we describe a novel variant of COG6 gene found in homozygosity in a Moroccan patient with severe presentation of COG6-CDG (OMIM #614576). We compared the phenotype of our patient with other previously reported COG6-CDG cases. Common features in COG6-CDG are facial dysmorphism, growth retardation, microcephaly, developmental disability, liver or gastrointestinal disease, recurrent infections, hypohidrosis/hyperthermia. In addition to these phenotypic features, our patient exhibited a disorder of sexual differentiation, which has rarely been reported in COG6-CDG. We hypothesize that the severe COG6 gene mutation interferes with glycosylation of a disintegrin and metalloprotease family members, inhibiting the correct gonadal distal tip cells migration, fundamental for the genitalia morphogenesis. This report broadens the genetic and phenotypic spectrum of COG6-CDG and provides further supportive evidence that COG6-CDG can present as a disorder of sexual differentiation.


Subject(s)
Abnormalities, Multiple/genetics , Adaptor Proteins, Vesicular Transport/genetics , Craniofacial Abnormalities/genetics , Disorders of Sex Development/genetics , Muscular Atrophy/genetics , Sexual Development/genetics , Abnormalities, Multiple/physiopathology , Codon, Nonsense/genetics , Congenital Disorders of Glycosylation/complications , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/physiopathology , Craniofacial Abnormalities/complications , Craniofacial Abnormalities/physiopathology , Disorders of Sex Development/complications , Disorders of Sex Development/physiopathology , Genetic Predisposition to Disease , Golgi Apparatus/genetics , Homozygote , Humans , Infant , Infant, Newborn , Karyotype , Male , Microcephaly/complications , Microcephaly/genetics , Microcephaly/physiopathology , Muscular Atrophy/complications , Muscular Atrophy/physiopathology , Phenotype
6.
J Inherit Metab Dis ; 43(6): 1370-1381, 2020 11.
Article in English | MEDLINE | ID: mdl-32852845

ABSTRACT

Congenital disorders of glycosylation (CDG) are a growing group of inborn metabolic disorders with multiorgan presentation. SLC39A8-CDG is a severe subtype caused by biallelic mutations in the manganese transporter SLC39A8, reducing levels of this essential cofactor for many enzymes including glycosyltransferases. The current diagnostic standard for disorders of N-glycosylation is the analysis of serum transferrin. Exome and Sanger sequencing were performed in two patients with severe neurodevelopmental phenotypes suggestive of CDG. Transferrin glycosylation was analyzed by high-performance liquid chromatography (HPLC) and isoelectric focusing in addition to comprehensive N-glycome analysis using matrix-assisted laser desorption ionization time of flight (MALDI-TOF) mass spectrometry (MS). Atomic absorption spectroscopy was used to quantify whole blood manganese levels. Both patients presented with a severe, multisystem disorder, and a complex neurological phenotype. Magnetic resonance imaging (MRI) revealed a Leigh-like syndrome with bilateral T2 hyperintensities of the basal ganglia. In patient 1, exome sequencing identified the previously undescribed homozygous variant c.608T>C [p.F203S] in SLC39A8. Patient 2 was found to be homozygous for c.112G>C [p.G38R]. Both individuals showed a reduction of whole blood manganese, though transferrin glycosylation was normal. N-glycome using MALDI-TOF MS identified an increase of the asialo-agalactosylated precursor N-glycan A2G1S1 and a decrease in bisected structures. In addition, analysis of heterozygous CDG-allele carriers identified similar but less severe glycosylation changes. Despite its reliance as a clinical gold standard, analysis of transferrin glycosylation cannot be categorically used to rule out SLC39A8-CDG. These results emphasize that SLC39A8-CDG presents as a spectrum of dysregulated glycosylation, and MS is an important tool for identifying deficiencies not detected by conventional methods.


Subject(s)
Basal Ganglia/physiopathology , Cation Transport Proteins/genetics , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/physiopathology , Adolescent , Cation Transport Proteins/deficiency , Child , Child, Preschool , Chromatography, High Pressure Liquid , Female , Glycosylation , Humans , Infant , Magnetic Resonance Imaging , Male , Manganese/metabolism , Mass Spectrometry , Phenotype , Transferrin/analysis , Exome Sequencing , Young Adult
7.
Clin Genet ; 98(4): 402-407, 2020 10.
Article in English | MEDLINE | ID: mdl-32683677

ABSTRACT

COG6-congenital disorder of glycosylation (COG6-CDG) is caused by biallelic mutations in COG6. To-date, 12 variants causing COG6-CDG in less than 20 patients have been reported. Using whole exome sequencing we identified two siblings with a novel homozygous deletion of 26 bp in COG6, creating a splicing variant (c.518_540 + 3del) and a shift in the reading frame. The phenotype of COG6-CDG includes growth and developmental retardation, microcephaly, liver and gastrointestinal disease, hypohydrosis and recurrent infections. We report two patients with novel phenotypic features including bowel malrotation and ambiguous genitalia, directing attention to the role of glycoprotein metabolism in the causation of disorders of sex development (DSD). Searching the glycomic literature, we identified 14 CDGs including males with DSD, a feature not previously accentuated. This study broadens the genetic and phenotypic spectrum of COG6-CDG and calls for increasing awareness to the central role of glycosylation processes in development of human sex and genitalia.


Subject(s)
Adaptor Proteins, Vesicular Transport/genetics , Congenital Disorders of Glycosylation/genetics , Disorders of Sex Development/genetics , Mixed Function Oxygenases/genetics , Congenital Disorders of Glycosylation/mortality , Congenital Disorders of Glycosylation/physiopathology , Disorders of Sex Development/mortality , Disorders of Sex Development/physiopathology , Female , Glycosylation , Homozygote , Humans , Infant, Newborn , Male , Mutation/genetics , Phenotype , Sequence Deletion/genetics , Siblings , Exome Sequencing
8.
J Mother Child ; 24(2): 14-20, 2020 Nov 10.
Article in English | MEDLINE | ID: mdl-33554500

ABSTRACT

This review discusses a selection of congenital disorders of glycosylation that show peculiar features, such as an unusual presentation, different phenotypes, a novel biochemical/genetic mechanism, a relatively high frequency or a relatively efficient treatment.


Subject(s)
Congenital Disorders of Glycosylation/complications , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/physiopathology , Symptom Assessment , Humans , Mutation , Phenotype
9.
Orphanet J Rare Dis ; 14(1): 148, 2019 06 19.
Article in English | MEDLINE | ID: mdl-31217022

ABSTRACT

BACKGROUND: NGLY1 deficiency is a rare autosomal recessive disorder caused by loss in enzymatic function of NGLY1, a peptide N-glycanase that has been shown to play a role in endoplasmic reticulum associated degradation (ERAD). ERAD dysfunction has been implicated in other well-described proteinopathies, such as Alzheimer's disease, Parkinson's disease, and Huntington's disease. The classical clinical tetrad includes developmental delay, hypolacrima, transiently elevated transaminases, and hyperkinetic movement disorders. The musculoskeletal system is also commonly affected, but the orthopaedic phenotype has been incompletely characterized. Best practices for orthopaedic clinical care have not been elucidated and considerable variability has resulted from this lack of evidence base. Our study surveyed patients enrolled in an international registry for NGLY1 deficiency in order to characterize the orthopaedic manifestations, sequelae, and management. RESULTS: Our findings, encompassing the largest cohort for NGLY1 deficiency to date, detail levels of motor milestone achievement; physical exam findings; fracture rates/distribution; frequency of motor skill regression; non-pharmacologic and non-procedural interventions; pharmacologic therapies; and procedural interventions experienced by 29 participants. Regarding the orthopaedic phenotype, at time of survey response, we found that over 40% of patients experienced motor skill regression from their peak. Over 80% of patients had at least one orthopaedic diagnosis, and nearly two-thirds of the total had two or more. More than half of patients older than 6 years had sustained a fracture. Related to orthopaedic non-medical management, we found that 93 and 79% of patients had utilized physical therapy and non-operative orthoses, respectively. In turn, the vast majority took at least one medication (including for bone health and antispasmodic therapy). Finally, nearly half of patients had undergone an invasive procedure. Of those older than 6 years, two-thirds had one or more procedures. Stratification of these analyses by sex revealed distinctive differences in disease natural history and clinical management course. CONCLUSIONS: These findings describing the orthopaedic natural history and standard of care in patients with NGLY1 deficiency can facilitate diagnosis, inform prognosis, and guide treatment recommendations in an evidence-based manner. Furthermore, the methodology is notable for its partnership with a disease-specific advocacy organization and may be generalizable to other rare disease populations. This study fills a void in the existing literature for this population and this methodology offers a precedent upon which future studies for rare diseases can build.


Subject(s)
Congenital Disorders of Glycosylation/pathology , Musculoskeletal System/pathology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Adolescent , Adult , Child , Child, Preschool , Congenital Disorders of Glycosylation/physiopathology , Evidence-Based Medicine/methods , Female , Humans , Infant , Male , Musculoskeletal System/physiopathology , Orthopedics , Rare Diseases , Young Adult
10.
Neuromuscul Disord ; 29(4): 282-289, 2019 04.
Article in English | MEDLINE | ID: mdl-30737079

ABSTRACT

The congenital disorders of glycosylation are a group of clinically and biochemically heterogeneous diseases characterized by multisystem involvement due to glycosylation defect of protein and lipid. Here we report a 49-year-old man with exercise-induced fatigue and pain of muscle, tachypnea, cleft palate and bifid uvula. Exercise induced elevation of serum creatine kinase (CK), ammonia and lactic acid was recorded. The abnormal levels of myoglobin, CK-MB and LDH as well as S-T elevation in electrocardiogram were observed in repeated hospitalization recordings. Electromyography showed myopathic damage. Repetitive nerve stimulation test of low rates showed decrement in the left deltoid muscle. He was identified with a novel homozygous frameshift variant in Phosphoglucomutase type 1 gene (c.405delT p.N135Kfs*9) by whole exome sequencing. Muscle biopsy exhibited minimal variation in fiber size without abnormal glycogen accumulation. Compared with controls', the patient's sample showed no signal at ∼61 kDa using N- or C-terminus antibody of Phosphoglucomutase type 1 in western blotting. A signal at ∼20 kDa was detected in patient using N-terminus antibody. Immunofluorescence revealed trace expression of C-terminus and a much lower expression of N-terminus on the sarcolemma than normal. Our findings indicate that c.405delT encodes a truncated protein with abnormal distribution and expression in skeletal muscle. In conclusion, genes associated with congenital disorders of glycosylation should be analyzed in patients with maxillofacial dysplasia, exertional weakness, cardiac involvement and exercise-induced-ammoniemia, without glycogen storage in skeletal muscle.


Subject(s)
Congenital Disorders of Glycosylation , Phosphoglucomutase/genetics , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/metabolism , Congenital Disorders of Glycosylation/pathology , Congenital Disorders of Glycosylation/physiopathology , Genetic Association Studies , Homozygote , Humans , Male , Middle Aged , Mutation , Pedigree , Exome Sequencing
11.
Genet Med ; 21(5): 1181-1188, 2019 05.
Article in English | MEDLINE | ID: mdl-30293989

ABSTRACT

PURPOSE: PMM2-CDG is the most common congenital disorder of glycosylation (CDG), which presents with either a neurologic or multisystem phenotype. Little is known about the longitudinal evolution. METHODS: We performed data analysis on PMM2-CDG patients' clinical features according to the Nijmegen CDG severity score and laboratory data. Seventy-five patients (28 males) were followed up from 11.0 ± 6.91 years for an average of 7.4 ± 4.5 years. RESULTS: On a group level, there was no significant evolution in overall clinical severity. There was some improvement in mobility and communication, liver and endocrine function, and strabismus and eye movements. Educational achievement and thyroid function worsened in some patients. Overall, the current clinical function, the system-specific involvement, and the current clinical assessment remained unchanged. On follow-up there was improvement of biochemical variables with (near) normalization of activated partial thromboplastin time (aPTT), factor XI, protein C, antithrombin, thyroid stimulating hormone, and liver transaminases. CONCLUSION: PMM2-CDG patients show a spontaneous biochemical improvement and stable clinical course based on the Nijmegen CDG severity score. This information is crucial for the definition of endpoints in clinical trials.


Subject(s)
Congenital Disorders of Glycosylation/epidemiology , Congenital Disorders of Glycosylation/physiopathology , Phosphotransferases (Phosphomutases)/deficiency , Adolescent , Child , Child, Preschool , Disease Progression , Female , Follow-Up Studies , Humans , Male , Phenotype , Young Adult
12.
Mitochondrion ; 38: 6-16, 2018 01.
Article in English | MEDLINE | ID: mdl-28750948

ABSTRACT

Mitochondrial respiratory chain (RC) diseases and congenital disorders of glycosylation (CDG) share extensive clinical overlap but are considered to have distinct cellular pathophysiology. Here, we demonstrate that an essential physiologic connection exists between cellular N-linked deglycosylation capacity and mitochondrial function. Following identification of altered muscle and liver mitochondrial amount and function in two children with a CDG subtype caused by NGLY1 deficiency, we evaluated mitochondrial physiology in NGLY1 disease human fibroblasts, and in NGLY1-knockout mouse embryonic fibroblasts and C. elegans. Across these distinct evolutionary models of cytosolic NGLY1 deficiency, a consistent disruption of mitochondrial physiology was present involving modestly reduced mitochondrial content with more pronounced impairment of mitochondrial membrane potential, increased mitochondrial matrix oxidant burden, and reduced cellular respiratory capacity. Lentiviral rescue restored NGLY1 expression and mitochondrial physiology in human and mouse fibroblasts, confirming that NGLY1 directly influences mitochondrial function. Overall, cellular deglycosylation capacity is shown to be a significant factor in mitochondrial RC disease pathogenesis across divergent evolutionary species.


Subject(s)
Congenital Disorders of Glycosylation/pathology , Congenital Disorders of Glycosylation/physiopathology , Fibroblasts/pathology , Fibroblasts/physiology , Mitochondria/pathology , Mitochondria/physiology , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Animals , Caenorhabditis elegans , Cell Respiration , Cells, Cultured , Child, Preschool , Electron Transport , Female , Gene Knockout Techniques , Genetic Complementation Test , Humans , Male , Membrane Potential, Mitochondrial , Mice, Knockout
13.
Am J Med Genet A ; 173(9): 2428-2434, 2017 Sep.
Article in English | MEDLINE | ID: mdl-28816422

ABSTRACT

Congenital disorders of glycosylation (CDGs) are a group of genetic diseases caused by mutations in genes that are necessary for the addition of oligosaccharides to acceptor proteins or lipids. An early step in this process requires dolichol kinase (DK) to catalyze the formation of dolichyl phosphate, which acts as a membrane anchor for initial attachment of sugar residues that are subsequently built up to oligosaccharides and transferred to acceptor proteins and lipids for further processing. Biallelic mutations in DOLK, the gene for DK, result in human in a CDG with variable symptoms, ranging from nonsyndromic dilated cardiomypopathy to severe multiorgan involvement. We report two female siblings with novel compound heterozygous mutations in DOLK: c.951C>A (p.Tyr317Ter) and c.1558A>G (p.Thr520Ala). Both patients presented in the neonatal period with severe ichthyosis, unusual distal digital constrictions and dilated cardiomyopathy which resulted in death. Histology of the skin showed lipid droplet accumulation in the stratum corneum and keratinocytes, which suggests defective epidermal lipid metabolism. These patients represent an earlier and more severe form of DOLK-CDG (CDG-1m) with a striking presentation at birth that expands the known phenotypic spectrum.


Subject(s)
Cardiomyopathy, Dilated/genetics , Congenital Disorders of Glycosylation/genetics , Phosphotransferases (Alcohol Group Acceptor)/deficiency , Cardiomyopathy, Dilated/complications , Cardiomyopathy, Dilated/physiopathology , Congenital Disorders of Glycosylation/physiopathology , Female , Humans , Ichthyosis/complications , Ichthyosis/genetics , Ichthyosis/physiopathology , Infant , Infant, Newborn , Lipid Metabolism/genetics , Mutation , Phenotype , Phosphotransferases (Alcohol Group Acceptor)/genetics , Siblings
14.
Rev. neurol. (Ed. impr.) ; 65(4): 161-176, 16 ago., 2017. ilus, tab, graf
Article in Spanish | IBECS | ID: ibc-166092

ABSTRACT

Desde la descripción de Engel del primer caso de miastenia congénita en 1977 y el hallazgo en 1995 del primer gen patógeno, el conocimiento de los síndromes miasténicos congénitos se ha ido desarrollando, y se han descrito la base patógena, sus características clínicas, las correlaciones fenotipo-genotipo establecidas y su abordaje terapéutico. En este grupo de enfermedades se altera el margen de seguridad de la transmisión neuromuscular por distintos mecanismos: en la síntesis o almacenamiento de los quantum de acetilcolina en las vesículas sinápticas, en la liberación de acetilcolina en el nervio terminal mediada por calcio o en la eficiencia de la cuanta liberada para generar una despolarización postsináptica. Su conocimiento ha permitido establecer distintas estrategias terapéuticas. En esta revisión se describen las principales actualizaciones de estos síndromes: los genes descritos que clasifican un 50% de los casos, su clasificación actual basándose en la localización de las proteínas que alteran la transmisión neuromuscular, incluyendo un nuevo grupo de miastenias congénitas, los trastornos de la glicosilación, las principales claves diagnósticas y el abordaje terapéutico de este grupo de pacientes infradiagnosticados (AU)


Since Engel reported the first case of congenital myasthenia in 1977 and the first pathogenic gene was found in 1995, knowledge about congenital myasthenic syndromes has continued to grow. Over the years, the pathogenic basis, its clinical features, the phenotype-genotype correlations that have been established and its therapeutic management have all been described. In this group of diseases the safety margin of neuromuscular transmission is altered by different mechanisms: in the synthesis or storage of acetylcholine quanta in the synaptic vesicles, in the calcium-mediated release of acetylcholine in the nerve terminal or in the efficiency of the quantum released to generate a post-synaptic depolarisation. Increased knowledge about them has enabled a number of different therapeutic strategies to be established. In this review the main updates on these syndromes are reported, including: the genes described as classifying 50% of cases, their current classification based on the localisation of the proteins that alter neuromuscular transmission, including a new group of congenital myasthenias, glycosylation disorders, the main key diagnoses and the therapeutic management of this group of under-diagnosed patients (AU)


Subject(s)
Humans , Myasthenic Syndromes, Congenital/classification , Congenital Disorders of Glycosylation/physiopathology , Myasthenia Gravis, Neonatal/genetics , Phenotype , Genotype , Mutation/genetics , Neuromuscular Junction/physiopathology
15.
Neurology ; 89(7): 657-664, 2017 Aug 15.
Article in English | MEDLINE | ID: mdl-28733338

ABSTRACT

OBJECTIVE: To describe the presentation and identify the cause of a new clinical phenotype, characterized by early severe neurodegeneration with myopathic and myasthenic features. METHODS: This case study of 5 patients from 3 families includes clinical phenotype, serial MRI, electrophysiologic testing, muscle biopsy, and full autopsy. Genetic workup included whole exome sequencing and segregation analysis of the likely causal mutation. RESULTS: All 5 patients showed severe muscular hypotonia, progressive cerebral atrophy, and therapy-refractory epilepsy. Three patients had congenital contractures. All patients died during their first year of life. In 2 of our patients, electrophysiologic testing showed abnormal decrement, but treatment with pyridostigmine led only to temporary improvement. Causative mutations in ALG14 were identified in all patients. The mutation c.220 G>A (p.Asp74Asn) was homozygous in 2 patients and heterozygous in the other 3 patients. Additional heterozygous mutations were c.422T>G (p.Val141Gly) and c.326G>A (p.Arg109Gln). In all cases, parents were found to be heterozygous carriers. None of the identified variants has been described previously. CONCLUSIONS: We report a genetic syndrome combining myasthenic features and severe neurodegeneration with therapy-refractory epilepsy. The underlying cause is a glycosylation defect due to mutations in ALG14. These cases broaden the phenotypic spectrum associated with ALG14 congenital disorders of glycosylation as previously only isolated myasthenia has been described.


Subject(s)
Cerebrum/pathology , Congenital Disorders of Glycosylation , Epilepsy , Muscle Weakness , N-Acetylglucosaminyltransferases/genetics , Atrophy/pathology , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/pathology , Congenital Disorders of Glycosylation/physiopathology , Epilepsy/genetics , Epilepsy/pathology , Epilepsy/physiopathology , Fatal Outcome , Female , Humans , Infant , Male , Muscle Weakness/genetics , Muscle Weakness/pathology , Muscle Weakness/physiopathology , Neurodegenerative Diseases , Pedigree , Phenotype , Syndrome
17.
Am J Med Genet A ; 173(6): 1620-1624, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28425223

ABSTRACT

Congenital disorders of glycosylation (CDG) are caused by defective glycosylation of proteins and lipids. PMM2-CDG is the most common subtype among the CDG. The severity of PMM2-CDG is variable. Patients often have a recognizable phenotype with neurological and multisystem symptoms that might cause early death. We report six patients from three families who are diagnosed with a clinically mild PMM2-CDG and have normal cognitive development. All these patients had delayed gross motor skills with mild-to-moderate neurological findings. Cerebellar hypoplasia was detected in all siblings for whom brain MRI was performed. In 5/6 children the Wechsler Intelligence Scale for Children (WISC) showed normal cognitive development with full scale IQ scores ranging from borderline to average. Four patients were diagnosed with PMM2-CDG at the age of 8 years or later as their neurological symptoms were quite mild and they had been able to participate in regular school programs. We report patients with p.Val231Met/p.Arg239Trp and p.Ile120Thr/p.Gly228Cys genotypes which may cause milder variants of PMM2-CDG.


Subject(s)
Cerebellum/abnormalities , Cognition/physiology , Congenital Disorders of Glycosylation/genetics , Nervous System Malformations/genetics , Phosphotransferases (Phosphomutases)/genetics , Adolescent , Cerebellum/diagnostic imaging , Cerebellum/physiopathology , Child , Child, Preschool , Congenital Disorders of Glycosylation/diagnostic imaging , Congenital Disorders of Glycosylation/physiopathology , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Genotype , Humans , Magnetic Resonance Imaging , Male , Mutation , Nervous System Malformations/diagnostic imaging , Nervous System Malformations/physiopathology , Phenotype
18.
J Neurosci Res ; 95(1-2): 362-370, 2017 01 02.
Article in English | MEDLINE | ID: mdl-27870454

ABSTRACT

This Mini-Review describes sex differences in 66 quantitative characteristics of the brain and body measured in a community-based sample of 1,024 adolescents 12-18 years of age, members of the Saguenay Youth Study. Using an extensive phenotyping protocol, we have obtained measures in a number of domains, including brain structure, cognition, mental health, substance use, body composition, metabolism, cardiovascular reactivity, and life style. For each measure, we provide estimates of effect size (Cohen's d) and sex-specific correlations with age (Pearson R). In total 59 of the 66 characteristics showed sex differences (at a nominal P < 0.05), with small (32), medium-sized (13), and large (11) effects. Some, but not all, of these sex differences increase during adolescence; this appears to be the case mostly for anatomical and physiological measures. © 2016 Wiley Periodicals, Inc.


Subject(s)
Brain/pathology , Brain/physiopathology , Congenital Disorders of Glycosylation/pathology , Mannose-6-Phosphate Isomerase/deficiency , Sex Characteristics , Adolescent , Age Factors , Child , Congenital Disorders of Glycosylation/physiopathology , Humans
19.
Am J Med Genet A ; 170(12): 3165-3171, 2016 12.
Article in English | MEDLINE | ID: mdl-27480077

ABSTRACT

Increasing numbers of congenital disorders of glycosylation (CDG) have been reported recently resulting in an expansion of the phenotypes associated with this group of disorders. SRD5A3 codes for polyprenol reductase which converts polyprenol to dolichol. This is a major pathway for dolichol biosynthesis for N-glycosylation, O-mannosylation, C-mannosylation, and GPI anchor synthesis. We present the features of five individuals (three children and two adults) with mutations in SRD5A3 focusing on the variable eye and skin involvement. We compare that to 13 affected individuals from the literature including five adults allowing us to delineate the features that may develop over time with this disorder including kyphosis, retinitis pigmentosa, and cataracts. © 2016 Wiley Periodicals, Inc.


Subject(s)
3-Oxo-5-alpha-Steroid 4-Dehydrogenase/genetics , Congenital Disorders of Glycosylation/genetics , Eye/physiopathology , Membrane Proteins/genetics , Skin/physiopathology , Adult , Child , Congenital Disorders of Glycosylation/physiopathology , Dolichols/metabolism , Female , Glycosylation , Homozygote , Humans , Male , Mutation , Phenotype , Tretinoin/analogs & derivatives , Tretinoin/metabolism
20.
Dis Model Mech ; 9(5): 513-27, 2016 05 01.
Article in English | MEDLINE | ID: mdl-26940433

ABSTRACT

Congenital disorders of glycosylation (CDGs) constitute a rapidly growing family of human diseases resulting from heritable mutations in genes driving the production and modification of glycoproteins. The resulting symptomatic hypoglycosylation causes multisystemic defects that include severe neurological impairments, revealing a particularly critical requirement for tightly regulated glycosylation in the nervous system. The most common CDG, CDG-Ia (PMM2-CDG), arises from phosphomannomutase type 2 (PMM2) mutations. Here, we report the generation and characterization of the first Drosophila CDG-Ia model. CRISPR-generated pmm2-null Drosophila mutants display severely disrupted glycosylation and early lethality, whereas RNAi-targeted knockdown of neuronal PMM2 results in a strong shift in the abundance of pauci-mannose glycan, progressive incoordination and later lethality, closely paralleling human CDG-Ia symptoms of shortened lifespan, movement impairments and defective neural development. Analyses of the well-characterized Drosophila neuromuscular junction (NMJ) reveal synaptic glycosylation loss accompanied by defects in both structural architecture and functional neurotransmission. NMJ synaptogenesis is driven by intercellular signals that traverse an extracellular synaptomatrix and are co-regulated by glycosylation and matrix metalloproteinases (MMPs). Specifically, trans-synaptic signaling by the Wnt protein Wingless (Wg) depends on the heparan sulfate proteoglycan (HSPG) co-receptor Dally-like protein (Dlp), which is regulated by synaptic MMP activity. Loss of synaptic MMP2, Wg ligand, Dlp co-receptor and downstream trans-synaptic signaling occurs with PMM2 knockdown. Taken together, this Drosophila CDG disease model provides a new avenue for the dissection of cellular and molecular mechanisms underlying neurological impairments and is a means by which to discover and test novel therapeutic treatment strategies.


Subject(s)
Congenital Disorders of Glycosylation/enzymology , Congenital Disorders of Glycosylation/pathology , Drosophila Proteins/metabolism , Drosophila/enzymology , Phosphotransferases (Phosphomutases)/deficiency , Synapses/enzymology , Animals , Congenital Disorders of Glycosylation/physiopathology , Disease Models, Animal , Down-Regulation , Extracellular Matrix/metabolism , Glycoproteins/metabolism , Glycosylation , Longevity , Movement , Neuromuscular Junction/metabolism , Oligosaccharides/metabolism , Phosphotransferases (Phosphomutases)/metabolism , Polysaccharides/metabolism , Posture , Presynaptic Terminals/metabolism , Signal Transduction , Synaptic Transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...