Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 93
Filtrar
1.
Mol Brain ; 14(1): 91, 2021 06 13.
Artigo em Inglês | MEDLINE | ID: mdl-34120625

RESUMO

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.


Assuntos
Defeitos Congênitos da Glicosilação/fisiopatologia , Atividade Motora/fisiologia , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/deficiência , Acetilglucosamina/análogos & derivados , Animais , Defeitos Congênitos da Glicosilação/enzimologia , Modelos Animais de Doenças , Terapia Genética , Vetores Genéticos/administração & dosagem , Gliose/complicações , Gliose/patologia , Humanos , Inflamação/patologia , Injeções Intraventriculares , Neurônios/patologia , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/genética , Ratos , Ratos Transgênicos , Transgenes
2.
Mol Genet Metab ; 131(1-2): 135-146, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33342467

RESUMO

Phosphoglucomutase 1 deficiency is a congenital disorder of glycosylation (CDG) with multiorgan involvement affecting carbohydrate metabolism, N-glycosylation and energy production. The metabolic management consists of dietary D-galactose supplementation that ameliorates hypoglycemia, hepatic dysfunction, endocrine anomalies and growth delay. Previous studies suggest that D-galactose administration in juvenile patients leads to more significant and long-lasting effects, stressing the urge of neonatal diagnosis (0-6 months of age). Here, we detail the early clinical presentation of PGM1-CDG in eleven infantile patients, and applied the modified Beutler test for screening of PGM1-CDG in neonatal dried blood spots (DBSs). All eleven infants presented episodic hypoglycemia and elevated transaminases, along with cleft palate and growth delay (10/11), muscle involvement (8/11), neurologic involvement (5/11), cardiac defects (2/11). Standard dietary measures for suspected lactose intolerance in four patients prior to diagnosis led to worsening of hypoglycemia, hepatic failure and recurrent diarrhea, which resolved upon D-galactose supplementation. To investigate possible differences in early vs. late clinical presentation, we performed the first systematic literature review for PGM1-CDG, which highlighted respiratory and gastrointestinal symptoms as significantly more diagnosed in neonatal age. The modified Butler-test successfully identified PGM1-CDG in DBSs from seven patients, including for the first time Guthrie cards from newborn screening, confirming the possibility of future inclusion of PGM1-CDG in neonatal screening programs. In conclusion, severe infantile morbidity of PGM1-CDG due to delayed diagnosis could be prevented by raising awareness on its early presentation and by inclusion in newborn screening programs, enabling early treatments and galactose-based metabolic management.


Assuntos
Defeitos Congênitos da Glicosilação/genética , Doença de Depósito de Glicogênio/sangue , Hipoglicemia/genética , Fosfoglucomutase/sangue , Fissura Palatina/sangue , Fissura Palatina/complicações , Fissura Palatina/genética , Defeitos Congênitos da Glicosilação/sangue , Defeitos Congênitos da Glicosilação/complicações , Defeitos Congênitos da Glicosilação/enzimologia , Teste em Amostras de Sangue Seco , Feminino , Doença de Depósito de Glicogênio/enzimologia , Doença de Depósito de Glicogênio/genética , Humanos , Hipoglicemia/sangue , Hipoglicemia/complicações , Lactente , Recém-Nascido , Masculino , Triagem Neonatal , Fenótipo , Fosfoglucomutase/genética
3.
BMC Med Genet ; 21(1): 135, 2020 06 23.
Artigo em Inglês | MEDLINE | ID: mdl-32576142

RESUMO

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.


Assuntos
Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/genética , Mutação/genética , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/deficiência , Sequência de Aminoácidos , Sequência de Bases , Encéfalo/diagnóstico por imagem , Encéfalo/patologia , Feminino , Heterozigoto , Humanos , Lactente , Recém-Nascido , Imageamento por Ressonância Magnética , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/química , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/genética
4.
J Inherit Metab Dis ; 43(4): 671-693, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32266963

RESUMO

Mannose phosphate isomerase-congenital disorder of glycosylation (MPI-CDG) deficiency is a rare subtype of congenital disorders of protein N-glycosylation. It is characterised by deficiency of MPI caused by pathogenic variants in MPI gene. The manifestation of MPI-CDG is different from other CDGs as the patients suffer dominantly from gastrointestinal and hepatic involvement whereas they usually do not present intellectual disability or neurological impairment. It is also one of the few treatable subtypes of CDGs with proven effect of oral mannose. This article covers a complex review of the literature and recommendations for the management of MPI-CDG with an emphasis on the clinical aspect of the disease. A team of international experts elaborated summaries and recommendations for diagnostics, differential diagnosis, management, and treatment of each system/organ involvement based on evidence-based data and experts' opinions. Those guidelines also reveal more questions about MPI-CDG which need to be further studied.


Assuntos
Defeitos Congênitos da Glicosilação/diagnóstico , Defeitos Congênitos da Glicosilação/terapia , Manose-6-Fosfato Isomerase/deficiência , Defeitos Congênitos da Glicosilação/enzimologia , Consenso , Gerenciamento Clínico , Humanos , Manose-6-Fosfato Isomerase/genética , Guias de Prática Clínica como Assunto
5.
Folia Biol (Praha) ; 65(3): 134-141, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31638560

RESUMO

Steroid 5α-reductase type 3 congenital disorder of glycosylation (SRD5A3-CDG) is a severe metabolic disease manifesting as muscle hypotonia, developmental delay, cerebellar ataxia and ocular symptoms; typically, nystagmus and optic disc pallor. Recently, early onset retinal dystrophy has been reported as an additional feature. In this study, we summarize ocular phenotypes and SRD5A3 variants reported to be associated with SRD5A3-CDG. We also describe in detail the ophthalmic findings in a 12-year-old Czech child harbouring a novel homozygous variant, c.436G>A, p.(Glu146Lys) in SRD5A3. The patient was reviewed for congenital nystagmus and bilateral optic neuropathy diagnosed at 13 months of age. Examination by spectral domain optical coherence tomography and fundus autofluorescence imaging showed clear signs of retinal dystrophy not recognized until our investigation. Best corrected visual acuity was decreased to 0.15 and 0.16 in the right and left eye, respectively, with a myopic refractive error of -3.0 dioptre sphere (DS) / -2.5 dioptre cylinder (DC) in the right and -3.0 DS / -3.0 DC in the left eye. The proband also had optic head nerve drusen, which have not been previously observed in this syndrome.


Assuntos
3-Oxo-5-alfa-Esteroide 4-Desidrogenase/genética , Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/genética , Olho/patologia , Proteínas de Membrana/genética , Mutação/genética , 3-Oxo-5-alfa-Esteroide 4-Desidrogenase/química , Sequência de Aminoácidos , Sequência de Bases , Criança , Feminino , Homozigoto , Humanos , Masculino , Proteínas de Membrana/química , Linhagem , Fenótipo
6.
J Inherit Metab Dis ; 42(5): 998-1007, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31077402

RESUMO

Patients with phosphoglucomutase (PGM1) deficiency, a congenital disorder of glycosylation (CDG) suffer from multiple disease phenotypes. Midline cleft defects are present at birth. Overtime, additional clinical phenotypes, which include severe hypoglycemia, hepatopathy, growth retardation, hormonal deficiencies, hemostatic anomalies, frequently lethal, early-onset of dilated cardiomyopathy and myopathy emerge, reflecting the central roles of the enzyme in (glycogen) metabolism and glycosylation. To delineate the pathophysiology of the tissue-specific disease phenotypes, we constructed a constitutive Pgm2 (mouse ortholog of human PGM1)-knockout (KO) mouse model using CRISPR-Cas9 technology. After multiple crosses between heterozygous parents, we were unable to identify homozygous life births in 78 newborn pups (P = 1.59897E-06), suggesting an embryonic lethality phenotype in the homozygotes. Ultrasound studies of the course of pregnancy confirmed Pgm2-deficient pups succumb before E9.5. Oral galactose supplementation (9 mg/mL drinking water) did not rescue the lethality. Biochemical studies of tissues and skin fibroblasts harvested from heterozygous animals confirmed reduced Pgm2 enzyme activity and abundance, but no change in glycogen content. However, glycomics analyses in serum revealed an abnormal glycosylation pattern in the Pgm2+/- animals, similar to that seen in PGM1-CDG.


Assuntos
Defeitos Congênitos da Glicosilação/tratamento farmacológico , Galactose/administração & dosagem , Genes Letais , Fosfoglucomutase/deficiência , Animais , Animais Recém-Nascidos , Defeitos Congênitos da Glicosilação/complicações , Defeitos Congênitos da Glicosilação/enzimologia , Feminino , Glicosilação , Heterozigoto , Homozigoto , Hipoglicemia/complicações , Masculino , Camundongos , Camundongos Knockout , Doenças Musculares/complicações , Doenças Musculares/patologia , Fenótipo
7.
Physiol Rep ; 7(3): e13979, 2019 02.
Artigo em Inglês | MEDLINE | ID: mdl-30740912

RESUMO

Homozygous mutations in NGLY1 were recently found to cause a condition characterized by a complex neurological syndrome, hypo- or alacrimia, and elevated liver transaminases. For yet unknown reasons, mortality is increased in patients with this condition. NGLY1 encodes the cytosolic enzyme N-glycanase 1, which is responsible for the deglycosylation of misfolded N-glycosylated proteins. Disruption of this process is hypothesized to lead to an accumulation of misfolded proteins in the cytosol. Here, we describe the disease course of a girl with a homozygous mutation in NGLY1, namely c.1837del (p.Gln613 fs). In addition to the previously described symptoms, at the age of 8 she presented with recurrent infections and hyperpigmentation, and, subsequently, a diagnosis of primary adrenal insufficiency was made. There are no previous reports describing adrenal insufficiency in such patients. We postulate that patients with NGLY1 deficiency may develop adrenal insufficiency as a consequence of impaired proteostasis, and the accompanying proteotoxic stress-induced cell death, through defective Nrf1 function. We recommend an annual evaluation of adrenal function in all patients with NGLY1 mutations in order to prevent unnecessary deaths.


Assuntos
Insuficiência Adrenal/genética , Defeitos Congênitos da Glicosilação/genética , Mutação , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/deficiência , Deficiências na Proteostase/genética , Proteostase/genética , Insuficiência Adrenal/diagnóstico , Insuficiência Adrenal/enzimologia , Criança , Defeitos Congênitos da Glicosilação/complicações , Defeitos Congênitos da Glicosilação/diagnóstico , Defeitos Congênitos da Glicosilação/enzimologia , Feminino , Predisposição Genética para Doença , Homozigoto , Humanos , Peptídeo-N4-(N-acetil-beta-glucosaminil) Asparagina Amidase/genética , Fenótipo , Prognóstico , Deficiências na Proteostase/diagnóstico , Deficiências na Proteostase/enzimologia
8.
J Biol Chem ; 293(52): 19957-19973, 2018 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-30389790

RESUMO

Glucosidase I (GI) removes the outermost glucose from protein-linked Glc3Man9GlcNAc2 (G3M9) in the endoplasmic reticulum (ER). Individuals with congenital disorders of glycosylation MOGS-CDG bear mutations in the GI-encoding gene (gls1). Although GI absence has been reported to produce lethality in Schizosaccharomyces pombe yeasts, here we obtained two viable Δgls1 mutants, one with a very sick but not lethal phenotype (Δgls1-S) and the other with a healthier one (Δgls1-H). The sick strain displayed only G3M9 as an ER protein-linked oligosaccharide, whereas the healthier strain had both G3M9 and Man9GlcNAc2 The lipid-linked oligosaccharide patterns of the two strains revealed that the most abundantly formed glycans were G3M9 in Δgls1-S and Glc2Man9GlcNAc2 in Δgls1-H, suggesting reduced Alg10p glucosyltransferase activity in the Δgls1-H strain. A mutation in the alg10+ gene was indeed observed in this strain. Our results indicated that abrogated G3M9 deglucosylation was responsible for the severe defects observed in Δgls1-S cells. Further studies disclosed that the defects could not be ascribed to disruption of glycoprotein entrance into calnexin-folding cycles, inhibition of the oligosaccharyltransferase by transfer reaction products, or reduced proteasomal degradation of misfolded glycoproteins. Lack of triglucosylated glycoprotein deglucosylation neither significantly prevented glycan elongation in the Golgi nor modified the overall cell wall monosaccharide composition. Nevertheless, it resulted in a distorted cell wall and in the absence of underlying ER membranes. Furthermore, Golgi expression of human endomannosidase partially restored normal growth in Δgls1-S cells. We propose that accumulation of G3M9-bearing glycoproteins is toxic and at least partially responsible for defects observed in MOGS-CDG.


Assuntos
Defeitos Congênitos da Glicosilação , Deleção de Genes , Modelos Biológicos , Proteínas de Schizosaccharomyces pombe , Schizosaccharomyces , alfa-Glucosidases , Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/genética , Retículo Endoplasmático/enzimologia , Retículo Endoplasmático/genética , Glucosiltransferases/genética , Glucosiltransferases/metabolismo , Glicosilação , Humanos , Oligossacarídeos/genética , Oligossacarídeos/metabolismo , Schizosaccharomyces/enzimologia , Schizosaccharomyces/genética , Proteínas de Schizosaccharomyces pombe/genética , Proteínas de Schizosaccharomyces pombe/metabolismo , alfa-Glucosidases/genética , alfa-Glucosidases/metabolismo
9.
PLoS One ; 12(12): e0189629, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-29261720

RESUMO

The most frequent disorder of glycosylation, PMM2-CDG, is caused by a deficiency of phosphomannomutase activity. In humans two paralogous enzymes exist, both of them require mannose 1,6-bis-phosphate or glucose 1,6-bis-phosphate as activators, but only phospho-mannomutase1 hydrolyzes bis-phosphate hexoses. Mutations in the gene encoding phosphomannomutase2 are responsible for PMM2-CDG. Although not directly causative of the disease, the role of the paralogous enzyme in the disease should be clarified. Phosphomannomutase1 could have a beneficial effect, contributing to mannose 6-phosphate isomerization, or a detrimental effect, hydrolyzing the bis-phosphate hexose activator. A pivotal role in regulating mannose-1phosphate production and ultimately protein glycosylation might be played by inosine monophosphate that enhances the phosphatase activity of phosphomannomutase1. In this paper we analyzed human phosphomannomutases by conventional enzymatic assays as well as by novel techniques such as 31P-NMR and thermal shift assay. We characterized a triple mutant of phospomannomutase1 that retains mutase and phosphatase activity, but is unable to bind inosine monophosphate.


Assuntos
Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/genética , Inosina Monofosfato/farmacologia , Mutação/genética , Fosfotransferases (Fosfomutases)/deficiência , Sequência de Aminoácidos , Difosfonatos/farmacologia , Ativação Enzimática/efeitos dos fármacos , Ensaios Enzimáticos , Estabilidade Enzimática/efeitos dos fármacos , Humanos , Ligantes , Espectroscopia de Ressonância Magnética , Simulação de Acoplamento Molecular , Fosfotransferases (Fosfomutases)/química , Fosfotransferases (Fosfomutases)/genética , Alinhamento de Sequência , Temperatura
10.
Orphanet J Rare Dis ; 11(1): 84, 2016 06 24.
Artigo em Inglês | MEDLINE | ID: mdl-27343064

RESUMO

BACKGROUND: Type I congenital disorders of glycosylation (CDG-I) are mostly complex multisystemic diseases associated with hypoglycosylated serum glycoproteins. A subgroup harbour mutations in genes necessary for the biosynthesis of the dolichol-linked oligosaccharide (DLO) precursor that is essential for protein N-glycosylation. Here, our objective was to identify the molecular origins of disease in such a CDG-Ix patient presenting with axial hypotonia, peripheral hypertonia, enlarged liver, micropenis, cryptorchidism and sensorineural deafness associated with hypo glycosylated serum glycoproteins. RESULTS: Targeted sequencing of DNA revealed a splice site mutation in intron 5 and a non-sense mutation in exon 4 of the dehydrodolichol diphosphate synthase gene (DHDDS). Skin biopsy fibroblasts derived from the patient revealed ~20 % residual DHDDS mRNA, ~35 % residual DHDDS activity, reduced dolichol-phosphate, truncated DLO and N-glycans, and an increased ratio of [2-(3)H]mannose labeled glycoprotein to [2-(3)H]mannose labeled DLO. Predicted truncated DHDDS transcripts did not complement rer2-deficient yeast. SiRNA-mediated down-regulation of DHDDS in human hepatocellular carcinoma HepG2 cells largely mirrored the biochemical phenotype of cells from the patient. The patient also harboured the homozygous ALG6(F304S) variant, which does not cause CDG but has been reported to be more frequent in PMM2-CDG patients with severe/fatal disease than in those with moderate presentations. WES did not reveal other strong candidate causal genes. CONCLUSIONS: We describe a patient presenting with severe multisystem disease associated with DHDDS deficiency. As retinitis pigmentosa is the only clinical sign in previously reported cases, this report broadens the spectrum of phenotypes associated with this condition.


Assuntos
Alquil e Aril Transferases/metabolismo , Defeitos Congênitos da Glicosilação/enzimologia , Cromatografia em Camada Fina , Defeitos Congênitos da Glicosilação/sangue , Defeitos Congênitos da Glicosilação/metabolismo , Dolicóis/análogos & derivados , Dolicóis/metabolismo , Éxons/genética , Glicoproteínas/sangue , Glicoproteínas/química , Glicoproteínas/metabolismo , Glicosilação , Células Hep G2 , Humanos , Recém-Nascido , Masculino , Mutação , Oligossacarídeos/metabolismo , Polissacarídeos/metabolismo , RNA Interferente Pequeno/genética , Pele/metabolismo
11.
Dis Model Mech ; 9(5): 513-27, 2016 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-26940433

RESUMO

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.


Assuntos
Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/patologia , Proteínas de Drosophila/metabolismo , Drosophila/enzimologia , Fosfotransferases (Fosfomutases)/deficiência , Sinapses/enzimologia , Animais , Defeitos Congênitos da Glicosilação/fisiopatologia , Modelos Animais de Doenças , Regulação para Baixo , Matriz Extracelular/metabolismo , Glicoproteínas/metabolismo , Glicosilação , Longevidade , Movimento , Junção Neuromuscular/metabolismo , Oligossacarídeos/metabolismo , Fosfotransferases (Fosfomutases)/metabolismo , Polissacarídeos/metabolismo , Postura , Terminações Pré-Sinápticas/metabolismo , Transdução de Sinais , Transmissão Sináptica
12.
Orphanet J Rare Dis ; 10: 138, 2015 Oct 26.
Artigo em Inglês | MEDLINE | ID: mdl-26502900

RESUMO

BACKGROUND: Phosphomannomutase deficiency (PMM2-CDG) is the most frequent congenital disorder of glycosylation. The cerebellum is nearly always affected in PMM2-CDG patients, a cerebellar atrophy progression is observed, and cerebellar dysfunction is their main daily functional limitation. Different therapeutic agents are under development, and clinical evaluation of drug candidates will require a standardized score of cerebellar dysfunction. We aim to assess the validity of the International Cooperative Ataxia Rating Scale (ICARS) in children and adolescents with genetically confirmed PMM2-CDG deficiency. We compare ICARS results with the Nijmegen Pediatric CDG Rating Scale (NPCRS), neuroimaging, intelligence quotient (IQ) and molecular data. METHODS: Our observational study included 13 PMM2-CDG patients and 21 control subjects. Ethical permissions and informed consents were obtained. Three independent child neurologists rated PMM2-CDG patients and control subjects using the ICARS. A single clinician administered the NPCRS. All patients underwent brain MRI, and the relative diameter of the midsagittal vermis was measured. Psychometric evaluations were available in six patients. The Mann-Whitney U test was used to compare ICARS between patients and controls. To evaluate inter-observer agreement in patients' ICARS ratings, intraclass correlation coefficients (ICC) were calculated. ICARS internal consistency was evaluated using Cronbach's alpha. Spearman's rank correlation coefficient test was used to correlate ICARS with NPCRS, midsagittal vermis relative diameter and IQ. RESULTS: ICARS and ICARS subscores differed between patients and controls (p < 0.001). Interobserver agreement of ICARS was "almost perfect" (ICC = 0.99), with a "good" internal reliability (Cronbach's alpha = 0.72). ICARS was significantly correlated with the total NPCRS score (rs 0.90, p < 0.001). However, there was no agreement regarding categories of severity. Regarding neuroimaging, inverse correlations between ICARS and midsagittal vermis relative diameter (rs -0.85, p = 0.003) and IQ (rs -0.94, p = 0.005) were found. Patients bearing p.E93A, p.C241S or p.R162W mutations presented a milder phenotype. CONCLUSIONS: ICARS is a reliable instrument for assessment of PMM2-CDG patients, without significant inter-rater variability. Despite our limited sample size, the results show a good correlation between functional cerebellar assessment, IQ and neuroimaging. For the first a correlation between ICARS, neuroimaging and IQ in PMM2-CDG patients has been demonstrated.


Assuntos
Ataxia Cerebelar/diagnóstico , Cerebelo/patologia , Defeitos Congênitos da Glicosilação/diagnóstico , Fosfotransferases (Fosfomutases)/deficiência , Índice de Gravidade de Doença , Adolescente , Ataxia Cerebelar/enzimologia , Ataxia Cerebelar/genética , Criança , Pré-Escolar , Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/genética , Feminino , Humanos , Masculino , Fosfotransferases (Fosfomutases)/genética
13.
Clin Genet ; 87(1): 42-8, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-24720419

RESUMO

Deficiencies in glycosyltransferases, glycosidases or nucleotide-sugar transporters involved in protein glycosylation lead to congenital disorders of glycosylation (CDG), a group of genetic diseases mostly showing multisystem phenotype. Despite recent advances in the biochemical and molecular knowledge of these diseases, no effective therapy exists for most. Efforts are now being directed toward therapies based on identifying new targets, which would allow to treat specific patients in a personalized way. This work presents proof-of concept for the antisense RNA rescue of the Golgi-resident protein TMEM165, a gene involved in a new type of CDG with a characteristic skeletal phenotype. Using a functional in vitro splicing assay based on minigenes, it was found that the deep intronic change c.792+182G>A is responsible for the insertion of an aberrant exon, corresponding to an intronic sequence. Antisense morpholino oligonucleotide therapy targeted toward TMEM165 mRNA recovered normal protein levels in the Golgi apparatus of patient-derived fibroblasts. This work expands the application of antisense oligonucleotide-mediated pseudoexon skipping to the treatment of a Golgi-resident protein, and opens up a promising treatment option for this specific TMEM165-CDG.


Assuntos
Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/terapia , Éxons/genética , Terapia Genética/métodos , Proteínas de Membrana/genética , Mutagênese Insercional/genética , RNA Antissenso/genética , Análise de Variância , Antiporters , Proteínas de Transporte de Cátions , Primers do DNA/genética , DNA Complementar/genética , Fibroblastos , Complexo de Golgi/genética , Humanos , Immunoblotting , Técnicas In Vitro/métodos , Microscopia de Fluorescência , Oligorribonucleotídeos Antissenso/genética , Oligorribonucleotídeos Antissenso/uso terapêutico
14.
Mol Genet Metab ; 112(4): 275-9, 2014 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-24997537

RESUMO

We recently redefined phosphoglucomutase-1 deficiency not only as an enzyme defect, involved in normal glycogen metabolism, but also an inborn error of protein glycosylation. Phosphoglucomutase-1 is a key enzyme in glycolysis and glycogenesis by catalyzing in the bidirectional transfer of phosphate from position 1 to 6 on glucose. Glucose-1-P and UDP-glucose are closely linked to galactose metabolism. Normal PGM1 activity is important for effective glycolysis during fasting. Activated glucose and galactose are essential for normal protein glycosylation. The complex defect involving abnormal concentrations of activated sugars leads to hypoglycemia and two major phenotypic presentations, one with primary muscle involvement and the other with severe multisystem disease. The multisystem phenotype includes growth delay and malformations, like cleft palate or uvula, and liver, endocrine and heart function with possible cardiomyopathy. The patients have normal intelligence. Decreased transferrin galactosylation is a characteristic finding on mass spectrometry. Previous in vitro studies in patient fibroblasts showed an improvement of glycosylation on galactose supplements. Four patients with PGM1 deficiency have been trialed on d-galactose (compassionate use), and showed improvement of serum transferrin hypoglycosylation. There was a parallel improvement of liver function, endocrine abnormalities and a decrease in the frequency of hypoglycemic episodes. No side effects have been observed. Galactose supplementation didn't seem to resolve all clinical symptoms. Adding complex carbohydrates showed an additional clinical amelioration. Based on the available clinical data we suggest to consider the use of 0.5-1g/kg/day d-galactose and maximum 50 g/day oral galactose therapy in PGM1-CDG. The existing data on galactose therapy have to be viewed as preliminary observations. A prospective multicenter trial is ongoing to evaluate the efficacy and optimal d-galactose dose of galactose supplementation.


Assuntos
Defeitos Congênitos da Glicosilação/tratamento farmacológico , Galactose/uso terapêutico , Fosfoglucomutase/deficiência , Defeitos Congênitos da Glicosilação/complicações , Defeitos Congênitos da Glicosilação/enzimologia , Humanos , Hipoglicemia/complicações , Doenças Musculares/complicações , Doenças Musculares/patologia , Fenótipo , Fosfoglucomutase/metabolismo
15.
Doc Ophthalmol ; 128(2): 149-53, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24493206

RESUMO

INTRODUCTION: We present the ocular features including full-field electroretinography (ff-ERG) and spectral domain optical coherence tomography (SD-OCT) in a 14-month-old infant with congenital disorder of glycosylation type 1a (PMM2-CDG). METHODS AND RESULTS: An infant with failure to thrive, bilateral neurosensory hearing loss, cerebellar hypoplasia, and pericardial effusions was referred to ophthalmic genetics for evaluation. The patient had fix and follow vision, an intermittent esotropia, moderate myopia, a hypo pigmented macula, and mild attenuation of the retinal vasculature. Electroretinography showed severe reduction in both rod and cone-dependent responses with a negative waveform pattern. Handheld SD-OCT revealed severe attenuation of the outer retina throughout the macula, but with preservation of outer retinal structures in the fovea. CONCLUSION: PMM2-CDG is a rare congenital disorder for which both ff-ERG and SD-OCT were useful in demonstrating early changes in retinal architecture and function.


Assuntos
Defeitos Congênitos da Glicosilação/diagnóstico , Esotropia/diagnóstico , Nistagmo Patológico/diagnóstico , Fosfotransferases (Fosfomutases)/deficiência , Doenças Retinianas/diagnóstico , Cerebelo/anormalidades , Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/fisiopatologia , Deficiências do Desenvolvimento/diagnóstico , Eletrorretinografia , Esotropia/enzimologia , Esotropia/fisiopatologia , Feminino , Humanos , Lactente , Malformações do Sistema Nervoso/diagnóstico , Nistagmo Patológico/enzimologia , Nistagmo Patológico/fisiopatologia , Retina/fisiopatologia , Doenças Retinianas/enzimologia , Doenças Retinianas/fisiopatologia , Tomografia de Coerência Óptica
16.
BMC Med Genet ; 15: 10, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24433453

RESUMO

BACKGROUND: SRD5A3 is responsible for SRD5A3-CDG, a type of congenital disorder of glycosylation, and mutations have been reported in 15 children. All the mutations are recessive and truncating. CASE PRESENTATION: We present 2 brothers at the age of 38 and 40 years with an initial diagnosis of cerebellar ataxia. We found the candidate disease loci via linkage analysis using data from single nucleotide polymorphism genome scans and homozygous truncating mutation SRD5A3 p.W19X, which was previously reported in 3 unrelated children, by exome sequencing.Clinical investigations included physical and ocular examinations and blood tests. Severe ocular involvement with retinal bone spicule pigmentation and optic atrophy are the most prominent disabling clinical features of the disease. The serum transferrin isoelectric focusing (TIEF) pattern is abnormal in the patient investigated. CONCLUSION: Our patients are older, with later onset and milder clinical phenotypes than all patients with SRD5A3-CDG reported so far. They also have atypical ocular findings and variable phenotypes. Our findings widen the spectrum of phenotypes resulting from SRD5A3 mutations and the clinical variability of SRD5A3-CDG, and suggest screening for SRD5A3 mutations in new patients with at least a few of the clinical symptoms of SRD5A3-CDG.


Assuntos
3-Oxo-5-alfa-Esteroide 4-Desidrogenase/genética , Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/genética , Proteínas de Membrana/genética , Fenótipo , Adulto , Defeitos Congênitos da Glicosilação/patologia , Defeitos Congênitos da Glicosilação/fisiopatologia , Feminino , Humanos , Masculino , Mutação , Polimorfismo de Nucleotídeo Único , Irmãos
17.
Dis Model Mech ; 6(1): 95-105, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-22899857

RESUMO

Individuals with congenital disorders of glycosylation (CDG) have recessive mutations in genes required for protein N-glycosylation, resulting in multi-systemic disease. Despite the well-characterized biochemical consequences in these individuals, the underlying cellular defects that contribute to CDG are not well understood. Synthesis of the lipid-linked oligosaccharide (LLO), which serves as the sugar donor for the N-glycosylation of secretory proteins, requires conversion of fructose-6-phosphate to mannose-6-phosphate via the phosphomannose isomerase (MPI) enzyme. Individuals who are deficient in MPI present with bleeding, diarrhea, edema, gastrointestinal bleeding and liver fibrosis. MPI-CDG patients can be treated with oral mannose supplements, which is converted to mannose-6-phosphate through a minor complementary metabolic pathway, restoring protein glycosylation and ameliorating most symptoms, although liver disease continues to progress. Because Mpi deletion in mice causes early embryonic lethality and thus is difficult to study, we used zebrafish to establish a model of MPI-CDG. We used a morpholino to block mpi mRNA translation and established a concentration that consistently yielded 13% residual Mpi enzyme activity at 4 days post-fertilization (dpf), which is within the range of MPI activity detected in fibroblasts from MPI-CDG patients. Fluorophore-assisted carbohydrate electrophoresis detected decreased LLO and N-glycans in mpi morphants. These deficiencies resulted in 50% embryonic lethality by 4 dpf. Multi-systemic abnormalities, including small eyes, dysmorphic jaws, pericardial edema, a small liver and curled tails, occurred in 82% of the surviving larvae. Importantly, these phenotypes could be rescued with mannose supplementation. Thus, parallel processes in fish and humans contribute to the phenotypes caused by Mpi depletion. Interestingly, mannose was only effective if provided prior to 24 hpf. These data provide insight into treatment efficacy and the broader molecular and developmental abnormalities that contribute to disorders associated with defective protein glycosylation.


Assuntos
Defeitos Congênitos da Glicosilação/dietoterapia , Defeitos Congênitos da Glicosilação/enzimologia , Manose-6-Fosfato Isomerase/deficiência , Manose-6-Fosfato Isomerase/genética , Manose/administração & dosagem , Animais , Sequência de Bases , Defeitos Congênitos da Glicosilação/genética , Suplementos Nutricionais , Modelos Animais de Doenças , Técnicas de Silenciamento de Genes , Humanos , Manose-6-Fosfato Isomerase/antagonistas & inibidores , Camundongos , Morfolinos/administração & dosagem , Morfolinos/genética , Mutação , Fenótipo , Peixe-Zebra/genética , Peixe-Zebra/metabolismo
18.
Eur J Hum Genet ; 21(8): 844-9, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23249953

RESUMO

Congenital disorders of glycosylation (CDG) are a large group of recessive multisystem disorders caused by impaired protein or lipid glycosylation. The CDG-I subgroup is characterized by protein N-glycosylation defects originating in the endoplasmic reticulum. The genetic defect is known for 17 different CDG-I subtypes. Patients in the few reported DPAGT1-CDG families exhibit severe intellectual disability (ID), epilepsy, microcephaly, severe hypotonia, facial dysmorphism and structural brain anomalies. In this study, we report a non-consanguineous family with two affected adults presenting with a relatively mild phenotype consisting of moderate ID, epilepsy, hypotonia, aggressive behavior and balance problems. Exome sequencing revealed a compound heterozygous missense mutation, c.85A>T (p.I29F) and c.503T>C (p.L168P), in the DPAGT1 gene. The affected amino acids are located in the first and fifth transmembrane domains of the protein. Isoelectric focusing and high-resolution mass spectrometry analyses of serum transferrin revealed glycosylation profiles that are consistent with a CDG-I defect. Our results show that the clinical spectrum of DPAGT1-CDG is much broader than appreciated so far.


Assuntos
Defeitos Congênitos da Glicosilação/genética , Heterozigoto , Mutação de Sentido Incorreto , N-Acetilglucosaminiltransferases/genética , Adulto , Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/patologia , Análise Mutacional de DNA/métodos , Exoma/genética , Feminino , Glicosilação , Humanos , Masculino , Fenótipo , Transferrina/metabolismo
19.
Mol Biol Cell ; 23(21): 4175-87, 2012 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-22956764

RESUMO

Congenital disorder of glycosylation (PMM2-CDG) results from mutations in pmm2, which encodes the phosphomannomutase (Pmm) that converts mannose-6-phosphate (M6P) to mannose-1-phosphate (M1P). Patients have wide-spectrum clinical abnormalities associated with impaired protein N-glycosylation. Although it has been widely proposed that Pmm2 deficiency depletes M1P, a precursor of GDP-mannose, and consequently suppresses lipid-linked oligosaccharide (LLO) levels needed for N-glycosylation, these deficiencies have not been demonstrated in patients or any animal model. Here we report a morpholino-based PMM2-CDG model in zebrafish. Morphant embryos had developmental abnormalities consistent with PMM2-CDG patients, including craniofacial defects and impaired motility associated with altered motor neurogenesis within the spinal cord. Significantly, global N-linked glycosylation and LLO levels were reduced in pmm2 morphants. Although M1P and GDP-mannose were below reliable detection/quantification limits, Pmm2 depletion unexpectedly caused accumulation of M6P, shown earlier to promote LLO cleavage in vitro. In pmm2 morphants, the free glycan by-products of LLO cleavage increased nearly twofold. Suppression of the M6P-synthesizing enzyme mannose phosphate isomerase within the pmm2 background normalized M6P levels and certain aspects of the craniofacial phenotype and abrogated pmm2-dependent LLO cleavage. In summary, we report the first zebrafish model of PMM2-CDG and uncover novel cellular insights not possible with other systems, including an M6P accumulation mechanism for underglycosylation.


Assuntos
Defeitos Congênitos da Glicosilação/enzimologia , Defeitos Congênitos da Glicosilação/patologia , Neurogênese , Fosfotransferases (Fosfomutases)/metabolismo , Proteínas de Peixe-Zebra/metabolismo , Peixe-Zebra/metabolismo , Animais , Cartilagem/efeitos dos fármacos , Cartilagem/embriologia , Cartilagem/patologia , Forma Celular/efeitos dos fármacos , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrócitos/patologia , Defeitos Congênitos da Glicosilação/genética , Anormalidades Craniofaciais/embriologia , Anormalidades Craniofaciais/patologia , Modelos Animais de Doenças , Embrião não Mamífero/anormalidades , Embrião não Mamífero/efeitos dos fármacos , Embrião não Mamífero/enzimologia , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Glicosilação/efeitos dos fármacos , Lipopolissacarídeos/metabolismo , Manose-6-Fosfato Isomerase/metabolismo , Manosefosfatos/metabolismo , Morfolinos/farmacologia , Neurônios Motores/efeitos dos fármacos , Neurônios Motores/patologia , Movimento/efeitos dos fármacos , Neurogênese/efeitos dos fármacos , Fosfotransferases (Fosfomutases)/deficiência , Fosfotransferases (Fosfomutases)/genética , Crânio/anormalidades , Crânio/efeitos dos fármacos , Crânio/embriologia , Medula Espinal/efeitos dos fármacos , Medula Espinal/embriologia , Medula Espinal/patologia , Especificidade por Substrato/efeitos dos fármacos , Peixe-Zebra/embriologia , Peixe-Zebra/genética , Proteínas de Peixe-Zebra/deficiência , Proteínas de Peixe-Zebra/genética
20.
Mol Genet Metab ; 105(4): 681-3, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22386715

RESUMO

Glycoproteins are essential in the production, transport, storage and regulation of thyroid hormones. Altered glycosylation has a potential impact on thyroid function. Abnormal thyroid function tests have been described in patients with congenital disorders of glycosylation. We evaluated the reliability of biochemical markers and investigated thyroid function in 18 PMM2-CDG patients. We propose an expectative therapeutic approach for neonates with thyroid abnormalities in CDG.


Assuntos
Defeitos Congênitos da Glicosilação/diagnóstico , Defeitos Congênitos da Glicosilação/terapia , Neuraminidase/farmacologia , Fosfotransferases (Fosfomutases)/metabolismo , Adolescente , Adulto , Criança , Pré-Escolar , Defeitos Congênitos da Glicosilação/enzimologia , Feminino , Seguimentos , Glicosilação , Humanos , Lactente , Recém-Nascido , Estudos Longitudinais , Masculino , Estudos Retrospectivos , Testes de Função Tireóidea , Tireotropina/metabolismo , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA