Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48
Filtrar
Más filtros

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
J Biol Chem ; 300(4): 107121, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-38417795

RESUMEN

Cytosolic peptide:N-glycanase (PNGase/NGLY1 in mammals) catalyzes deglycosylation of N-glycans on glycoproteins. A genetic disorder caused by mutations in the NGLY1 gene leads to NGLY1 deficiency with symptoms including motor deficits and neurological problems. Effective therapies have not been established, though, a recent study used the administration of an adeno-associated viral vector expressing human NGLY1 to dramatically rescue motor functions in young Ngly1-/- rats. Thus, early therapeutic intervention may improve symptoms arising from central nervous system dysfunction, and assay methods for measuring NGLY1 activity in biological samples are critical for early diagnostics. In this study, we established an assay system for plate-based detection of endogenous NGLY1 activity using a FRET-based probe. Using this method, we revealed significant changes in NGLY1 activity in rat brains during aging. This novel assay offers reliable disease diagnostics and provides valuable insights into the regulation of PNGase/NGLY1 activity in diverse organisms under different stress conditions.


Asunto(s)
Trastornos Congénitos de Glicosilación , Transferencia Resonante de Energía de Fluorescencia , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa , Animales , Humanos , Masculino , Ratas , Envejecimiento/metabolismo , Encéfalo/metabolismo , Trastornos Congénitos de Glicosilación/diagnóstico , Transferencia Resonante de Energía de Fluorescencia/métodos , Células HEK293 , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia
2.
Am J Hum Genet ; 109(2): 345-360, 2022 02 03.
Artículo en Inglés | MEDLINE | ID: mdl-35045343

RESUMEN

Free oligosaccharides (fOSs) are soluble oligosaccharide species generated during N-glycosylation of proteins. Although little is known about fOS metabolism, the recent identification of NGLY1 deficiency, a congenital disorder of deglycosylation (CDDG) caused by loss of function of an enzyme involved in fOS metabolism, has elicited increased interest in fOS processing. The catabolism of fOSs has been linked to the activity of a specific cytosolic mannosidase, MAN2C1, which cleaves α1,2-, α1,3-, and α1,6-mannose residues. In this study, we report the clinical, biochemical, and molecular features of six individuals, including two fetuses, with bi-allelic pathogenic variants in MAN2C1; the individuals are from four different families. These individuals exhibit dysmorphic facial features, congenital anomalies such as tongue hamartoma, variable degrees of intellectual disability, and brain anomalies including polymicrogyria, interhemispheric cysts, hypothalamic hamartoma, callosal anomalies, and hypoplasia of brainstem and cerebellar vermis. Complementation experiments with isogenic MAN2C1-KO HAP1 cells confirm the pathogenicity of three of the identified MAN2C1 variants. We further demonstrate that MAN2C1 variants lead to accumulation and delay in the processing of fOSs in proband-derived cells. These results emphasize the involvement of MAN2C1 in human neurodevelopmental disease and the importance of fOS catabolism.


Asunto(s)
Quistes del Sistema Nervioso Central/genética , Trastornos Congénitos de Glicosilación/genética , Hamartoma/genética , Discapacidad Intelectual/genética , Oligosacáridos/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Polimicrogiria/genética , alfa-Manosidasa/genética , Adolescente , Alelos , Tronco Encefálico/metabolismo , Tronco Encefálico/patología , Línea Celular Tumoral , Quistes del Sistema Nervioso Central/metabolismo , Quistes del Sistema Nervioso Central/patología , Vermis Cerebeloso/metabolismo , Vermis Cerebeloso/patología , Niño , Preescolar , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/patología , Femenino , Feto , Glicosilación , Hamartoma/metabolismo , Hamartoma/patología , Humanos , Hipotálamo/metabolismo , Hipotálamo/patología , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Leucocitos/metabolismo , Leucocitos/patología , Masculino , Manosa/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Polimicrogiria/metabolismo , Polimicrogiria/patología , Lengua/metabolismo , Lengua/patología , alfa-Manosidasa/deficiencia
3.
PLoS Genet ; 18(6): e1010228, 2022 06.
Artículo en Inglés | MEDLINE | ID: mdl-35653343

RESUMEN

NGLY1 deficiency, a rare disease with no effective treatment, is caused by autosomal recessive, loss-of-function mutations in the N-glycanase 1 (NGLY1) gene and is characterized by global developmental delay, hypotonia, alacrima, and seizures. We used a Drosophila model of NGLY1 deficiency to conduct an in vivo, unbiased, small molecule, repurposing screen of FDA-approved drugs to identify therapeutic compounds. Seventeen molecules partially rescued lethality in a patient-specific NGLY1 deficiency model, including multiple serotonin and dopamine modulators. Exclusive dNGLY1 expression in serotonin and dopamine neurons, in an otherwise dNGLY1 deficient fly, was sufficient to partially rescue lethality. Further, genetic modifier and transcriptomic data supports the importance of serotonin signaling in NGLY1 deficiency. Connectivity Map analysis identified glycogen synthase kinase 3 (GSK3) inhibition as a potential therapeutic mechanism for NGLY1 deficiency, which we experimentally validated with TWS119, lithium, and GSK3 knockdown. Strikingly, GSK3 inhibitors and a serotonin modulator rescued size defects in dNGLY1 deficient larvae upon proteasome inhibition, suggesting that these compounds act through NRF1, a transcription factor that is regulated by NGLY1 and regulates proteasome expression. This study reveals the importance of the serotonin pathway in NGLY1 deficiency, and serotonin modulators or GSK3 inhibitors may be effective therapeutics for this rare disease.


Asunto(s)
Reposicionamiento de Medicamentos , Glucógeno Sintasa Quinasa 3 , Animales , Trastornos Congénitos de Glicosilación , Drosophila/genética , Drosophila/metabolismo , Humanos , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Complejo de la Endopetidasa Proteasomal/metabolismo , Enfermedades Raras , Serotonina/genética
4.
Biochem Biophys Res Commun ; 710: 149826, 2024 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-38581946

RESUMEN

Cytosolic peptide:N-glycanase (NGLY1, PNGase) is an enzyme that cleaves N-glycans from misfolded glycoproteins. In 2012, a human genetic disorder, NGLY1 deficiency, was first reported to be caused by mutations of the NGLY1 gene. Since then, there has been rapid progresses on NGLY1 biology, and gene therapy has been proposed as a promising therapeutic option for NGLY1 deficiency. While a plasma/urine biomarker has also been developed for this disease, detection of NGLY1 activity could be another viable option for early diagnosis of NGLY1 deficiency. Thus far, several in vitro and in cellulo NGLY1 assays have been reported, but those assay systems have several issues that must be addressed in order to develop an assay system compatible for routine clinical examination. Here, we show a facile, highly sensitive in vitro assay system that could be used to detect NGLY1 activity by utilizing its sequence editing function, i.e. conversion of glycosylated Asn into Asp, followed by a detection of newly generated epitope (HA)-tag by anti-HA antibody. Using this ELISA-based assay, we detected endogenous NGLY1 activity in as little as 2 µg of crude extract, which is the equivalent of 5 × 103 cells. Our system also detects NGLY1 activity from cells with compromised NGLY1 activity, such as iPS cells from patient samples. This assay system could be applied in future clinical examinations to achieve an early diagnosis of NGLY1 deficiency.


Asunto(s)
Trastornos Congénitos de Glicosilación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Humanos , Citosol/metabolismo , Glicosilación , Glicoproteínas/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética
5.
PLoS Genet ; 16(12): e1009258, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-33315951

RESUMEN

Mutations in human N-glycanase 1 (NGLY1) cause the first known congenital disorder of deglycosylation (CDDG). Patients with this rare disease, which is also known as NGLY1 deficiency, exhibit global developmental delay and other phenotypes including neuropathy, movement disorder, and constipation. NGLY1 is known to regulate proteasomal and mitophagy gene expression through activation of a transcription factor called "nuclear factor erythroid 2-like 1" (NFE2L1). Loss of NGLY1 has also been shown to impair energy metabolism, but the molecular basis for this phenotype and its in vivo consequences are not well understood. Using a combination of genetic studies, imaging, and biochemical assays, here we report that loss of NGLY1 in the visceral muscle of the Drosophila larval intestine results in a severe reduction in the level of AMP-activated protein kinase α (AMPKα), leading to energy metabolism defects, impaired gut peristalsis, failure to empty the gut, and animal lethality. Ngly1-/- mouse embryonic fibroblasts and NGLY1 deficiency patient fibroblasts also show reduced AMPKα levels. Moreover, pharmacological activation of AMPK signaling significantly suppressed the energy metabolism defects in these cells. Importantly, the reduced AMPKα level and impaired energy metabolism observed in NGLY1 deficiency models are not caused by the loss of NFE2L1 activity. Taken together, these observations identify reduced AMPK signaling as a conserved mediator of energy metabolism defects in NGLY1 deficiency and suggest AMPK signaling as a therapeutic target in this disease.


Asunto(s)
Trastornos Congénitos de Glicosilación/metabolismo , Proteínas de Drosophila/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Proteínas Quinasas/metabolismo , Quinasas de la Proteína-Quinasa Activada por el AMP , Animales , Células Cultivadas , Proteínas de Drosophila/genética , Drosophila melanogaster , Metabolismo Energético , Fibroblastos/metabolismo , Humanos , Ratones , Factor 1 Relacionado con NF-E2/metabolismo , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Proteínas Quinasas/genética , Transducción de Señal
6.
Hum Mol Genet ; 29(10): 1635-1647, 2020 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-32259258

RESUMEN

N-glycanase 1 (NGLY1) deficiency, an autosomal recessive disease caused by mutations in the NGLY1 gene, is characterized by developmental delay, hypolacrima or alacrima, seizure, intellectual disability, movement disorders and other neurological phenotypes. Because of few animal models that recapitulate these clinical signatures, the mechanisms of the onset of the disease and its progression are poorly understood, and the development of therapies is hindered. In this study, we generated the systemic Ngly1-deficient rodent model, Ngly1-/- rats, which showed developmental delay, movement disorder, somatosensory impairment and scoliosis. These phenotypes in Ngly1-/- rats are consistent with symptoms in human patients. In accordance with the pivotal role played by NGLY1 in endoplasmic reticulum-associated degradation processes, cleaving N-glycans from misfolded glycoproteins in the cytosol before they can be degraded by the proteasome, loss of Ngly1 led to accumulation of cytoplasmic ubiquitinated proteins, a marker of misfolded proteins in the neurons of the central nervous system of Ngly1-/- rats. Histological analysis identified prominent pathological abnormalities, including necrotic lesions, mineralization, intra- and extracellular eosinophilic bodies, astrogliosis, microgliosis and significant loss of mature neurons in the thalamic lateral and the medial parts of the ventral posterior nucleus and ventral lateral nucleus of Ngly1-/- rats. Axonal degradation in the sciatic nerves was also observed, as in human subjects. Ngly1-/- rats, which mimic the symptoms of human patients, will be a useful animal model for preclinical testing of therapeutic options and understanding the detailed mechanisms of NGLY1 deficiency.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Trastornos del Movimiento/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Animales , Sistema Nervioso Central/metabolismo , Sistema Nervioso Central/patología , Trastornos Congénitos de Glicosilación/metabolismo , Trastornos Congénitos de Glicosilación/patología , Modelos Animales de Enfermedad , Degradación Asociada con el Retículo Endoplásmico/genética , Enfermedades Hereditarias del Ojo , Técnicas de Inactivación de Genes , Glicosilación , Humanos , Discapacidad Intelectual/genética , Discapacidad Intelectual/metabolismo , Discapacidad Intelectual/patología , Enfermedades del Aparato Lagrimal , Trastornos del Movimiento/patología , Mutación/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Sistema Nervioso Periférico/metabolismo , Sistema Nervioso Periférico/patología , Fenotipo , Complejo de la Endopetidasa Proteasomal/genética , Ratas
7.
J Inherit Metab Dis ; 45(3): 571-583, 2022 05.
Artículo en Inglés | MEDLINE | ID: mdl-35243670

RESUMEN

We delineated the phenotypic spectrum of epilepsy in individuals with NGLY1 deficiency from an international cohort. We collected detailed clinical and electroencephalographic data from 29 individuals with bi-allelic (likely) pathogenic variants in NGLY1 as part of an ongoing prospective natural history study. Participants were evaluated in-person at a single center and/or remotely. Historical medical records were reviewed. Published cases were included for comprehensive phenotyping. Of 29 individuals (mean 11.4 years, range 3-27 years), 17 (58.6%) participants had a history of epilepsy. Seizure onset was in early childhood (mean 43 months, range 2 months to 19 years). The most common seizure types were myoclonic and atonic. Epilepsy course was variable, but 35.2% (6/17) of participants with epilepsy achieved seizure freedom. The most common medications included levetiracetam, valproate, lamotrigine, and clobazam. Electroencephalogram (EEGs) were abnormal in 80% (12/15) of participants with or without epilepsy, although encephalopathy was uncommon. There was a trend in neurodevelopmental outcomes that participants with epilepsy had more developmental delays. In summary, epilepsy is common in NGLY1 deficiency. Over half of the participants had a history of epilepsy and nearly all had EEG abnormalities indicating an increased risk of epilepsy. This work expands the electroclinical phenotype of NGLY1 deficiency and supports a high clinical suspicion for seizures. Some of the more common seizure types (epileptic spasms, myoclonic, and atonic seizures) can be subtle and require counseling to ensure early recognition and treatment to ensure the best possible outcomes. Despite transient liver enzyme abnormalities in this disorder, hepatically metabolized medications were well tolerated.


Asunto(s)
Epilepsia , Preescolar , Trastornos Congénitos de Glicosilación , Electroencefalografía , Epilepsia/tratamiento farmacológico , Epilepsia/genética , Humanos , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Fenotipo , Estudios Prospectivos , Convulsiones/genética
8.
Artículo en Inglés | MEDLINE | ID: mdl-33563880

RESUMEN

N-Glycanase 1 (NGLY1) deficiency is a congenital disorder caused by mutations in the NGLY1 gene. Because systemic Ngly1-/- mice with a C57BL/6 (B6) background are embryonically lethal, studies on the mechanism of NGLY1 deficiency using mice have been problematic. In this study, B6-Ngly1-/+ mice were crossed with Japanese wild mice-originated Japanese fancy mouse 1 (JF1) mice to produce viable F2 Ngly1-/- mice from (JF1×B6)F1 Ngly1-/+ mice. Systemic Ngly1-/- mice with a JF1 mouse background were also embryonically lethal. Hybrid F1 Ngly1-/- (JF1/B6F1) mice, however, showed developmental delay and motor dysfunction, similar to that in human patients. JF1/B6F1 Ngly1-/- mice showed increased levels of plasma and urinary aspartylglycosamine, a potential biomarker for NGLY1 deficiency. JF1/B6F1 Ngly1-/- mice are a useful isogenic animal model for the preclinical testing of therapeutic options and understanding the precise pathogenic mechanisms responsible for NGLY1 deficiency.


Asunto(s)
Trastornos Congénitos de Glicosilación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Acetilglucosamina/análogos & derivados , Acetilglucosamina/sangre , Acetilglucosamina/genética , Animales , Trastornos Congénitos de Glicosilación/sangre , Trastornos Congénitos de Glicosilación/genética , Modelos Animales de Enfermedad , Humanos , Ratones , Ratones Endogámicos C57BL , Mutación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/sangre , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética
9.
Hum Mol Genet ; 27(6): 1055-1066, 2018 03 15.
Artículo en Inglés | MEDLINE | ID: mdl-29346549

RESUMEN

Autosomal recessive loss-of-function mutations in N-glycanase 1 (NGLY1) cause NGLY1 deficiency, the only known human disease of deglycosylation. Patients present with developmental delay, movement disorder, seizures, liver dysfunction and alacrima. NGLY1 is a conserved cytoplasmic component of the Endoplasmic Reticulum Associated Degradation (ERAD) pathway. ERAD clears misfolded proteins from the ER lumen. However, it is unclear how loss of NGLY1 function impacts ERAD and other cellular processes and results in the constellation of problems associated with NGLY1 deficiency. To understand how loss of NGLY1 contributes to disease, we developed a Drosophila model of NGLY1 deficiency. Loss of NGLY1 function resulted in developmental delay and lethality. We used RNAseq to determine which processes are misregulated in the absence of NGLY1. Transcriptome analysis showed no evidence of ER stress upon NGLY1 knockdown. However, loss of NGLY1 resulted in a strong signature of NRF1 dysfunction among downregulated genes, as evidenced by an enrichment of genes encoding proteasome components and proteins involved in oxidation-reduction. A number of transcriptome changes also suggested potential therapeutic interventions, including dysregulation of GlcNAc synthesis and upregulation of the heat shock response. We show that increasing the function of both pathways rescues lethality. Together, transcriptome analysis in a Drosophila model of NGLY1 deficiency provides insight into potential therapeutic approaches.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Acetilglucosamina/biosíntesis , Animales , Discapacidades del Desarrollo/metabolismo , Modelos Animales de Enfermedad , Proteínas de Drosophila/genética , Drosophila melanogaster/genética , Retículo Endoplásmico/metabolismo , Degradación Asociada con el Retículo Endoplásmico/genética , Glicosilación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Complejo de la Endopetidasa Proteasomal/metabolismo , Convulsiones/metabolismo , Análisis de Secuencia de ARN/métodos , Transcriptoma/genética
10.
Hum Mol Genet ; 27(17): 3029-3045, 2018 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-29878199

RESUMEN

Genomics methodologies have significantly improved elucidation of Mendelian disorders. The combination with high-throughput functional-omics technologies potentiates the identification and confirmation of causative genetic variants, especially in singleton families of recessive inheritance. In a cohort of 99 individuals with abnormal Golgi glycosylation, 47 of which being unsolved, glycomics profiling was performed of total plasma glycoproteins. Combination with whole-exome sequencing in 31 cases revealed a known genetic defect in 15 individuals. To identify additional genetic factors, hierarchical clustering of the plasma glycomics data was done, which indicated a subgroup of four patients that shared a unique glycomics signature of hybrid type N-glycans. In two siblings, compound heterozygous mutations were found in SLC10A7, a gene of unknown function in human. These included a missense mutation that disrupted transmembrane domain 4 and a mutation in a splice acceptor site resulting in skipping of exon 9. The two other individuals showed a complete loss of SLC10A7 mRNA. The patients' phenotype consisted of amelogenesis imperfecta, skeletal dysplasia, and decreased bone mineral density compatible with osteoporosis. The patients' phenotype was mirrored in SLC10A7 deficient zebrafish. Furthermore, alizarin red staining of calcium deposits in zebrafish morphants showed a strong reduction in bone mineralization. Cell biology studies in fibroblasts of affected individuals showed intracellular mislocalization of glycoproteins and a defect in post-Golgi transport of glycoproteins to the cell membrane. In contrast to yeast, human SLC10A7 localized to the Golgi. Our combined data indicate an important role for SLC10A7 in bone mineralization and transport of glycoproteins to the extracellular matrix.


Asunto(s)
Enfermedades del Desarrollo Óseo/etiología , Calcificación Fisiológica , Trastornos Congénitos de Glicosilación/complicaciones , Genómica , Glicómica , Mutación , Transportadores de Anión Orgánico Sodio-Dependiente/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Simportadores/genética , Adulto , Animales , Enfermedades del Desarrollo Óseo/metabolismo , Enfermedades del Desarrollo Óseo/patología , Células Cultivadas , Estudios de Cohortes , Exoma , Femenino , Fibroblastos/metabolismo , Fibroblastos/patología , Glicosilación , Aparato de Golgi/metabolismo , Aparato de Golgi/patología , Humanos , Lactante , Masculino , Transportadores de Anión Orgánico Sodio-Dependiente/metabolismo , Linaje , Fenotipo , Transporte de Proteínas , Simportadores/metabolismo , Adulto Joven , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Pez Cebra/metabolismo
11.
BMC Med Genet ; 21(1): 135, 2020 06 23.
Artículo en Inglés | MEDLINE | ID: mdl-32576142

RESUMEN

BACKGROUND: NGLY1-related congenital disorder of deglycosylation (NGLY1-CDDG) is a multisystemic neurodevelopmental disorder in which affected individuals show developmental delay, epilepsy, intellectual disability, abnormal liver function, and poor growth. This study presents a 10-month-old female infant with elevated liver transaminases, developmental delay, epilepsy (subclinical seizures), and constipation who possesses two compound heterozygous mutations in NGLY1. CASE PRESENTATION: The proband was admitted to the Department of Gastroenterology, Children's Hospital of Soochow University, with elevated liver transaminases. She had a history of intrauterine growth retardation and exhibited elevated transaminases, global developmental delay, seizures and light constipation during early infancy. Whole-exome sequencing (WES) and Sanger sequencing revealed two compound heterozygous mutations in NGLY1 that had been inherited in an autosomal recessive manner from her parents. One was a termination mutation, c.1168C > T (p.R390*), and the other was a missense mutation, c.1156G > T (p.D386Y). NGLY1-CDDG is a rare disorder, with a few dozen cases. The two mutations of this proband has not been previously identified. CONCLUSIONS: This study investigated a Chinese proband with NGLY1-CDDG born from healthy parents who was studied using WES and Sanger sequencing to identify the causative mutations. We identified two novel compound heterozygous mutations in NGLY1, c.1168C > T (p.R390*)/c.1156G > T (p.D386Y), which are probably causative of disease.


Asunto(s)
Trastornos Congénitos de Glicosilación/enzimología , Trastornos Congénitos de Glicosilación/genética , Mutación/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Secuencia de Aminoácidos , Secuencia de Bases , Encéfalo/diagnóstico por imagen , Encéfalo/patología , Femenino , Heterocigoto , Humanos , Lactante , Recién Nacido , Imagen por Resonancia Magnética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/química , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética
12.
J Hum Genet ; 65(4): 387-396, 2020 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-31965062

RESUMEN

NGLY1 deficiency is the first and only autosomal recessive congenital disorder of N-linked deglycosylation (NGLY1-CDDG). To date, no patients with NGLY1 deficiency has been reported from mainland China or East Asia in English literature. Here, we present six patients with a diagnosis of NGLY1-CDDG on the basis of clinical phenotype, genetic testing, and functional studies. We retrospectively analyzed clinical phenotypes and NGLY1 genotypes of six cases from four families. Informed consent was obtained for diagnosis and treatment. In-silico tools and in vitro enzyme activity assays were used to determine pathogenicity of NGLY1 varaints. All patients had typical features of NGLY1-CDDG, including global developmental delay, microcephaly, hypotonia, hypertransaminasemia, alacrimia, and feeding difficulty. Dysmorphic features found in our patients include flat nasal bridge, loose and hollow cheeks, short stature, malnutrition, and ptosis. Pachylosis could be a novel cutaneous feature that may be explained by lack of sweat. We found three novel variants, including one missense (c.982C > G/p.Arg328Gly), one splice site (c.1003+3A > G), and one frame-shift (c.1637-1652delCATCTTTTGCTTATAT/p.Ser546PhefsTer) variant. All mutations were predicted to be disease causing with in-silico prediction tools, and affected at least one feature of gene splicing. Protein modeling showed missense variants may affect covalent bonding within the protein structure, or interrupt active/binding amino-acid residues. In vitro studies indicated that proteins carrying missense variants (p.Arg328Gly and p.Tyr342Cys) lost the enzyme activity. We expanded clinical phenotype and genetic mutation spectrum of NGLY1-CDDG by reporting six cases, three novel variants, and novel clinical features from mainland China.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Discapacidades del Desarrollo/genética , Enfermedades Hereditarias del Ojo/genética , Trastornos de Alimentación y de la Ingestión de Alimentos/genética , Enfermedades del Aparato Lagrimal/genética , Microcefalia/genética , Hipotonía Muscular/genética , Mutación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Trastornos Congénitos de Glicosilación/patología , Discapacidades del Desarrollo/patología , Enfermedades Hereditarias del Ojo/patología , Trastornos de Alimentación y de la Ingestión de Alimentos/patología , Femenino , Humanos , Lactante , Enfermedades del Aparato Lagrimal/patología , Masculino , Microcefalia/patología , Hipotonía Muscular/patología , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética
13.
Pediatr Transplant ; 24(8): e13808, 2020 12.
Artículo en Inglés | MEDLINE | ID: mdl-32790950

RESUMEN

Patients with defects in the ATP6AP1 gene have rarely been described. ATP6AP1-related disorders are a subtype of CDG, which result in enzyme deficiencies affecting multiple organ systems ranging from mild to life-threatening. Of the 13 patients described, all had hepatopathy, but this is the first case to be successfully transplanted. We describe two brothers who developed hyperbilirubinemia shortly after birth and progressed to liver failure, case 1 by 12 months of age, with successful transplant 2 years later, and case 2 by 4 months of age, who passed away while awaiting liver transplant. Both boys were found to have a new variant in the ATP6AP1 gene: c.932/p.Leu311Gln. Although the identified ATP6AP1 gene variant was classified as unknown significance at the time, both children's phenotypes fit with what has been described for ATP6AP1-related disorders. Therefore, this result appears to have been diagnostic for both boys. This rare type of CDG, X-linked immunodeficiency type 47 (OMIM #300972), particularly in patients who progress to liver failure requiring transplant, should be included on the differential of liver failure in infants and toddlers, and its gene should be added to the diagnostic workup for such cases.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Trastornos Congénitos de Glicosilación/cirugía , Fallo Hepático/genética , Trasplante de Hígado , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , ATPasas de Translocación de Protón Vacuolares/genética , Resultado Fatal , Humanos , Lactante , Masculino , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Fenotipo , Hermanos
14.
PLoS Genet ; 13(4): e1006696, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28426790

RESUMEN

The cytoplasmic peptide:N-glycanase (Ngly1 in mammals) is a de-N-glycosylating enzyme that is highly conserved among eukaryotes. It was recently reported that subjects harboring mutations in the NGLY1 gene exhibited severe systemic symptoms (NGLY1-deficiency). While the enzyme obviously has a critical role in mammals, its precise function remains unclear. In this study, we analyzed Ngly1-deficient mice and found that they are embryonic lethal in C57BL/6 background. Surprisingly, the additional deletion of the gene encoding endo-ß-N-acetylglucosaminidase (Engase), which is another de-N-glycosylating enzyme but leaves a single GlcNAc at glycosylated Asn residues, resulted in the partial rescue of the lethality of the Ngly1-deficient mice. Additionally, we also found that a change in the genetic background of C57BL/6 mice, produced by crossing the mice with an outbred mouse strain (ICR) could partially rescue the embryonic lethality of Ngly1-deficient mice. Viable Ngly1-deficient mice in a C57BL/6 and ICR mixed background, however, showed a very severe phenotype reminiscent of the symptoms of NGLY1-deficiency subjects. Again, many of those defects were strongly suppressed by the additional deletion of Engase in the C57BL/6 and ICR mixed background. The defects observed in Ngly1/Engase-deficient mice (C57BL/6 background) and Ngly1-deficient mice (C57BL/6 and ICR mixed background) closely resembled some of the symptoms of patients with an NGLY1-deficiency. These observations strongly suggest that the Ngly1- or Ngly1/Engase-deficient mice could serve as a valuable animal model for studies related to the pathogenesis of the NGLY1-deficiency, and that cytoplasmic ENGase represents one of the potential therapeutic targets for this genetic disorder.


Asunto(s)
Enfermedades Genéticas Congénitas/genética , Manosil-Glicoproteína Endo-beta-N-Acetilglucosaminidasa/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Animales , Citoplasma/enzimología , Enfermedades Genéticas Congénitas/terapia , Glicosilación , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Modelos Animales , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/metabolismo , Eliminación de Secuencia/genética
15.
Pol J Pathol ; 71(1): 66-68, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32429657

RESUMEN

N-glycanase 1 deficiency is a congenital disorder of deglycosylation, which has been diagnosed in 27 patients, including 2 of them from Poland. The most characteristic symptoms include global developmental disability, hyperkinetic movement disorder, hypo-/alacrimia, and elevated serum transaminases. We reported on a patient in whom the liver biopsy done at the age of 3 years revealed the presence of steatosis, fibrosis, and an amorphous periodic acid-Schiff staining positive diastases-digested material in the cytoplasm.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Hígado/patología , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Preescolar , Humanos , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Polonia
16.
Postepy Biochem ; 66(1): 38-41, 2020 03 31.
Artículo en Polaco | MEDLINE | ID: mdl-33320481

RESUMEN

Together with the lysosomal storage diseases, N-glycanase 1 deficiency is a congenital disorder of deglycosylation, which has been diagnosed in 27 patients, including two of them from Poland. The pathogenesis remains unknown, however, the main role is attributed to the disturbed endoplasmic reticulum-associated protein degradation process. The most characteristic symptoms include global developmental disability, hyperkinetic movement disorder, hypo-/alacrimia, and elevated serum transaminases. Identification of pathogenic variants in the NGLY1 gene is required to confirm the diagnosis.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Trastornos Congénitos de Glicosilación/diagnóstico , Glicosilación , Humanos
17.
Hum Mutat ; 40(7): 938-951, 2019 07.
Artículo en Inglés | MEDLINE | ID: mdl-31067009

RESUMEN

ALG3-CDG is one of the very rare types of congenital disorder of glycosylation (CDG) caused by variants in the ER-mannosyltransferase ALG3. Here, we summarize the clinical, biochemical, and genetic data of four new ALG3-CDG patients, who were identified by a type I pattern of serum transferrin and the accumulation of Man5 GlcNAc2 -PP-dolichol in LLO analysis. Additional clinical symptoms observed in our patients comprise sensorineural hearing loss, right-descending aorta, obstructive cardiomyopathy, macroglossia, and muscular hypertonia. We add four new biochemically confirmed variants to the list of ALG3-CDG inducing variants: c.350G>C (p.R117P), c.1263G>A (p.W421*), c.1037A>G (p.N346S), and the intron variant c.296+4A>G. Furthermore, in Patient 1 an additional open-reading frame of 141 bp (AAGRP) in the coding region of ALG3 was identified. Additionally, we show that control cells synthesize, to a minor degree, a hybrid protein composed of the polypeptide AAGRP and ALG3 (AAGRP-ALG3), while in Patient 1 expression of this hybrid protein is significantly increased due to the homozygous variant c.160_196del (g.165C>T). By reviewing the literature and combining our findings with previously published data, we further expand the knowledge of this rare glycosylation defect.


Asunto(s)
Trastornos Congénitos de Glicosilación/genética , Manosiltransferasas/genética , Mutación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Animales , Células COS , Células Cultivadas , Preescolar , Chlorocebus aethiops , Femenino , Humanos , Lactante , Masculino , Sistemas de Lectura Abierta , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Polimorfismo de Nucleótido Simple
18.
Mol Genet Metab ; 127(4): 368-372, 2019 08.
Artículo en Inglés | MEDLINE | ID: mdl-31311714

RESUMEN

BACKGROUND: NGLY1-CDDG is a congenital disorder of deglycosylation caused by a defective peptide:N-glycanase (PNG). To date, all but one of the reported patients have been diagnosed through whole-exome or whole-genome sequencing, as no biochemical marker was available to identify this disease in patients. Recently, a potential urinary biomarker was reported, but the data presented suggest that this marker may be excreted intermittently. METHODS: In this study, we performed untargeted direct-infusion high-resolution mass spectrometry metabolomics in seven dried blood spots (DBS) from four recently diagnosed NGLY1-CDDG patients, to test for small-molecule biomarkers, in order to identify a potential diagnostic marker. Results were compared to 125 DBS of healthy controls and to 238 DBS of patients with other diseases. RESULTS: We identified aspartylglycosamine as the only significantly increased compound with a median Z-score of 4.8 (range: 3.8-8.5) in DBS of NGLY1-CDDG patients, compared to a median Z-score of -0.1 (range: -2.1-4.0) in DBS of healthy controls and patients with other diseases. DISCUSSION: The increase of aspartylglycosamine can be explained by lack of function of PNG. PNG catalyzes the cleavage of the proximal N-acetylglucosamine residue of an N-glycan from the asparagine residue of a protein, a step in the degradation of misfolded glycoproteins. PNG deficiency results in a single N-acetylglucosamine residue left attached to the asparagine residue which results in free aspartylglycosamine when the glycoprotein is degraded. Thus, we here identified aspartylglycosamine as the first potential small-molecule biomarker in DBS for NGLY1-CDDG, making a biochemical diagnosis for NGLY1-CDDG potentially feasible.


Asunto(s)
Acetilglucosamina/análogos & derivados , Trastornos Congénitos de Glicosilación/diagnóstico , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Acetilglucosamina/sangre , Adolescente , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Niño , Preescolar , Trastornos Congénitos de Glicosilación/sangre , Pruebas con Sangre Seca , Femenino , Humanos , Lactante , Masculino , Espectrometría de Masas , Mutación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/sangre
19.
Mol Genet Metab ; 124(1): 82-86, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29550355

RESUMEN

N-glycanase deficiency (NGLY1 deficiency, NGLY1-CDDG), the first autosomal recessive congenital disorder of N-linked deglycosylation (CDDG), is caused by pathogenic variants in NGLY1. The majority of affected individuals have been identified using exome or genome sequencing. To date, no reliable, clinically available biomarkers have been identified. Urine oligosaccharide analysis was included as part of a routine evaluation for possible biomarkers in patients with confirmed NGLY1-CDDG. During the qualitative review of oligosaccharide profiles by an experienced laboratory director an abnormal analyte with a proposed structure of Neu5Ac1Hex1GlcNAc1-Asn was identified in NGLY1-CDDG patient urine samples. The same species has been observed in profiles from individuals affected with aspartylglucosaminuria, although the complete spectra are not identical. Additional studies using tandem mass spectrometry confirmed the analyte's structure. In addition to the known NGLY1-CDDG patients identified by this analysis, a single case was identified in a population referred for clinical testing who subsequently had a diagnosis of NGLY1-CDDG confirmed by molecular testing. Urine oligosaccharide screening by MALDI-TOF MS can identify individuals with NGLY1-CDDG. In addition, this potential biomarker might also be used to monitor the effectiveness of therapeutic options as they become available.


Asunto(s)
Trastornos Congénitos de Glicosilación/diagnóstico , Oligosacáridos/orina , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Adolescente , Biomarcadores/orina , Niño , Preescolar , Trastornos Congénitos de Glicosilación/orina , Femenino , Humanos , Lactante , Masculino , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/aislamiento & purificación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/orina , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Espectrometría de Masas en Tándem , Adulto Joven
20.
Genet Med ; 19(2): 160-168, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-27388694

RESUMEN

PURPOSE: The cytosolic enzyme N-glycanase 1, encoded by NGLY1, catalyzes cleavage of the ß-aspartyl glycosylamine bond of N-linked glycoproteins, releasing intact N-glycans from proteins bound for degradation. In this study, we describe the clinical spectrum of NGLY1 deficiency (NGLY1-CDDG). METHODS: Prospective natural history protocol. RESULTS: In 12 individuals ages 2 to 21 years with confirmed, biallelic, pathogenic NGLY1 mutations, we identified previously unreported clinical features, including optic atrophy and retinal pigmentary changes/cone dystrophy, delayed bone age, joint hypermobility, and lower than predicted resting energy expenditure. Novel laboratory findings include low cerebral spinal fluid (CSF) total protein and albumin and unusually high antibody titers toward rubella and/or rubeola following vaccination. We also confirmed and further quantified previously reported findings noting that decreased tear production, transient transaminitis, small feet, a complex hyperkinetic movement disorder, and varying degrees of global developmental delay with relatively preserved socialization are the most consistent features. CONCLUSION: Our prospective phenotyping expands the clinical spectrum of NGLY1-CDDG, offers prognostic information, and provides baseline data for evaluating therapeutic interventions.Genet Med 19 2, 160-168.


Asunto(s)
Discapacidades del Desarrollo/genética , Glicoproteínas/genética , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/genética , Adolescente , Adulto , Albúminas/líquido cefalorraquídeo , Proteínas del Líquido Cefalorraquídeo/genética , Niño , Preescolar , Discapacidades del Desarrollo/fisiopatología , Femenino , Glicosilación , Humanos , Masculino , Mutación , Péptido-N4-(N-acetil-beta-glucosaminil) Asparagina Amidasa/deficiencia , Fenotipo , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA