Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 68
Filter
1.
Am J Hum Genet ; 108(6): 1040-1052, 2021 06 03.
Article in English | MEDLINE | ID: mdl-33964207

ABSTRACT

SLC37A4 encodes an endoplasmic reticulum (ER)-localized multitransmembrane protein required for transporting glucose-6-phosphate (Glc-6P) into the ER. Once transported into the ER, Glc-6P is subsequently hydrolyzed by tissue-specific phosphatases to glucose and inorganic phosphate during times of glucose depletion. Pathogenic variants in SLC37A4 cause an established recessive disorder known as glycogen storage disorder 1b characterized by liver and kidney dysfunction with neutropenia. We report seven individuals who presented with liver dysfunction multifactorial coagulation deficiency and cardiac issues and were heterozygous for the same variant, c.1267C>T (p.Arg423∗), in SLC37A4; the affected individuals were from four unrelated families. Serum samples from affected individuals showed profound accumulation of both high mannose and hybrid type N-glycans, while N-glycans in fibroblasts and undifferentiated iPSC were normal. Due to the liver-specific nature of this disorder, we generated a CRISPR base-edited hepatoma cell line harboring the c.1267C>T (p.Arg423∗) variant. These cells replicated the secreted abnormalities seen in serum N-glycosylation, and a portion of the mutant protein appears to relocate to a distinct, non-Golgi compartment, possibly ER exit sites. These cells also show a gene dosage-dependent alteration in the Golgi morphology and reduced intraluminal pH that may account for the altered glycosylation. In summary, we identify a recurrent mutation in SLC37A4 that causes a dominantly inherited congenital disorder of glycosylation characterized by coagulopathy and liver dysfunction with abnormal serum N-glycans.


Subject(s)
Antiporters/genetics , Congenital Disorders of Glycosylation/etiology , Endoplasmic Reticulum/pathology , Liver Diseases/complications , Monosaccharide Transport Proteins/genetics , Mutation , Adult , Child , Child, Preschool , Congenital Disorders of Glycosylation/pathology , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Genes, Dominant , Glycosylation , Humans , Infant , Infant, Newborn , Male , Pedigree
2.
J Clin Immunol ; 41(5): 958-966, 2021 07.
Article in English | MEDLINE | ID: mdl-33534079

ABSTRACT

Phosphoglucomutase 3 (PGM3) deficiency is a rare congenital disorder of glycosylation. Most of patients with autosomal recessive hypomorphic mutations in PGM3 encoding for phosphoglucomutase 3 present with eczema, skin and lung infections, elevated serum IgE, as well as neurological and skeletal features. A few PGM3-deficient patients suffer from a more severe disease with nearly absent T cells and severe skeletal dysplasia. We performed targeted next-generation sequencing on two kindred to identify the underlying genetic etiology of a severe combined immunodeficiency with developmental defect. We report here two novel homozygous missense variants (p.Gly359Asp and p.Met423Thr) in PGM3 identified in three patients from two unrelated kindreds with severe combined immunodeficiency, neurological impairment, and skeletal dysplasia. Both variants segregated with the disease in the two families. They were predicted to be deleterious by in silico analysis. PGM3 enzymatic activity was found to be severely impaired in primary fibroblasts and Epstein-Barr virus immortalized B cells from the kindred carrying the p.Met423Thr variant. Our findings support the pathogenicity of these two novel variants in severe PGM3 deficiency.


Subject(s)
Abnormalities, Multiple/genetics , Bone Diseases, Developmental/genetics , Limb Deformities, Congenital/genetics , Nervous System Diseases/genetics , Phosphoglucomutase/genetics , Severe Combined Immunodeficiency/genetics , Child, Preschool , Face/abnormalities , Female , Humans , Infant , Infant, Newborn , Male
3.
Hum Mutat ; 42(2): 142-149, 2021 02.
Article in English | MEDLINE | ID: mdl-33300232

ABSTRACT

Signal sequence receptor protein 4 (SSR4) is a subunit of the translocon-associated protein complex, which participates in the translocation of proteins across the endoplasmic reticulum membrane, enhancing the efficiency of N-linked glycosylation. Pathogenic variants in SSR4 cause a congenital disorder of glycosylation: SSR4-congenital disorders of glycosylation (CDG). We describe three SSR4-CDG boys and review the previously reported. All subjects presented with hypotonia, failure to thrive, developmental delay, and dysmorphic traits and showed a type 1 serum sialotransferrin profile, facilitating the diagnosis. Genetic confirmation of this X-linked CDG revealed one de novo hemizygous deletion, one maternally inherited deletion, and one de novo nonsense mutation of SSR4. The present subjects highlight the similarities with a connective tissue disorder (redundant skin, joint laxity, blue sclerae, and vascular tortuosity). The connective tissue problems are relevant, and require preventive rehabilitation measures. As an X-linked disorder, genetic counseling is essential.


Subject(s)
Calcium-Binding Proteins , Congenital Disorders of Glycosylation , Membrane Glycoproteins , Receptors, Cytoplasmic and Nuclear , Receptors, Peptide , Calcium-Binding Proteins/genetics , Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/pathology , Connective Tissue/pathology , Glycosylation , Humans , Male , Membrane Glycoproteins/genetics , Phenotype , Receptors, Cytoplasmic and Nuclear/genetics , Receptors, Peptide/genetics
4.
Environ Res ; 196: 110350, 2021 05.
Article in English | MEDLINE | ID: mdl-33144047

ABSTRACT

The skin is an essential barrier, protecting the body against the environment and its numerous pollutants. Several environmental pollutants are known to affect the skin, inducing premature aging through mechanisms including oxidative stress, inflammation, and impairment of skin functions. Even climate conditions can impact the skin. Therefore, using a Reconstructed Human Epidermis (RHE), we tested the effect of two samples of fine particulate matters (PM0.3-2.5 - one metals-rich sample and the other organic compounds-rich), two Volatile Organic Compounds mixtures (VOCs - from a solvent-based paint and a water-based paint) and Tobacco Smoke (TS). All pollutants affected cellular functionality, but to a lesser extent for the water-based paint VOC. This effect was enhanced when RHE were preconditioned for 2 h by a semi-dry airflow (45% relative humidity) before pollutants application, compared to preconditioning by a humid airflow (90% relative humidity). In the absence of preconditioning, IL-1α, IL-6, IL-8, and RANTES were almost systematically induced by pollutants. When RHE were preconditioned by a semi-dry or humid airflow before being subjected to pollutants, the increase of IL-1α, IL-8, and RANTES falls into two groups. Similarly to RHE not treated with pollutants, RHE treated with VOCs after preconditioning by a semi-dry airflow showed increased IL-1α, IL-8, and RANTES release. On the contrary, RHE treated with PM or TS after preconditioning by a semi-dry airflow show a lower increase in IL-1α, IL-8, and RANTES compared to preconditioning by a humid airflow. The effect of real environmental relative humidity conditions of the air, combined with acute exposure to various environmental pollutants, seemed to relate mainly to structural changes of the skin, determining the outcome of the inflammatory response depending on the physicochemical characteristics of pollutants.


Subject(s)
Air Pollutants , Environmental Pollutants , Volatile Organic Compounds , Air Pollutants/analysis , Air Pollutants/toxicity , Environmental Pollutants/toxicity , Humans , Humidity , Particulate Matter/analysis , Particulate Matter/toxicity , Volatile Organic Compounds/analysis , Volatile Organic Compounds/toxicity
5.
J Inherit Metab Dis ; 43(6): 1360-1369, 2020 11.
Article in English | MEDLINE | ID: mdl-33098580

ABSTRACT

Mannose phosphate isomerase MPI-CDG (formerly CDG-1b) is a potentially fatal inherited metabolic disease which is readily treatable with oral D-mannose. We retrospectively reviewed long-term outcomes of patients with MPI-CDG, all but one of whom were treated with D-mannose. Clinical, biological, and histological data were reviewed at diagnosis and on D-mannose treatment. Nine patients were diagnosed with MPI-CDG at a median age of 3 months. The presenting symptoms were diarrhea (n = 9), hepatomegaly (n = 9), hypoglycemia (n = 8), and protein loosing enteropathy (n = 7). All patients survived except the untreated one who died at 2 years of age. Oral D-mannose was started in eight patients at a median age of 7 months (mean 38 months), with a median follow-up on treatment of 14 years 9 months (1.5-20 years). On treatment, two patients developed severe portal hypertension, two developed venous thrombosis, and 1 displayed altered kidney function. Poor compliance with D-mannose was correlated with recurrence of diarrhea, thrombosis, and abnormal biological parameters including coagulation factors and transferrin profiles. Liver fibrosis persisted despite treatment, but two patients showed improved liver architecture during follow-up. This study highlights (i) the efficacy and safety of D-mannose treatment with a median follow-up on treatment of almost 15 years (ii) the need for life-long treatment (iii) the risk of relapse with poor compliance, (iii) the importance of portal hypertension screening (iv) the need to be aware of venous and renal complications in adulthood.


Subject(s)
Congenital Disorders of Glycosylation/drug therapy , Mannose-6-Phosphate Isomerase/deficiency , Mannose/administration & dosage , Mannose/adverse effects , Administration, Oral , Child , Child, Preschool , Female , Humans , Hypertension/etiology , Infant , Liver Cirrhosis/pathology , Male , Medication Adherence , Retrospective Studies , Transferrin/analysis , Treatment Outcome , Venous Thrombosis/etiology
6.
J Inherit Metab Dis ; 43(6): 1349-1359, 2020 11.
Article in English | MEDLINE | ID: mdl-32700771

ABSTRACT

Bikunin (Bkn) isoforms are serum chondroitin sulfate (CS) proteoglycans synthesized by the liver. They include two light forms, that is, the Bkn core protein and the Bkn linked to the CS chain (urinary trypsin inhibitor [UTI]), and two heavy forms, that is, pro-α-trypsin inhibitor and inter-α-trypsin inhibitor, corresponding to UTI esterified by one or two heavy chains glycoproteins, respectively. We previously showed that the Western-blot analysis of the light forms could allow the fast and easy detection of patients with linkeropathy, deficient in enzymes involved in the synthesis of the initial common tetrasaccharide linker of glycosaminoglycans. Here, we analyzed all serum Bkn isoforms in a context of congenital disorders of glycosylation (CDG) and showed very specific abnormal patterns suggesting potential interests for their screening and diagnosis. In particular, genetic deficiencies in V-ATPase (ATP6V0A2-CDG, CCDC115-CDG, ATP6AP1-CDG), in Golgi manganese homeostasis (TMEM165-CDG) and in the N-acetyl-glucosamine Golgi transport (SLC35A3-CDG) all share specific abnormal Bkn patterns. Furthermore, for each studied linkeropathy, we show that the light abnormal Bkn could be further in-depth characterized by two-dimensional electrophoresis. Moreover, besides being interesting as a specific biomarker of both CDG and linkeropathies, Bkn isoforms' analyses can provide new insights into the pathophysiology of the aforementioned diseases.


Subject(s)
Alpha-Globulins/metabolism , Antiporters/metabolism , Cation Transport Proteins/metabolism , Congenital Disorders of Glycosylation/metabolism , Golgi Apparatus/metabolism , Nucleotide Transport Proteins/metabolism , Biomarkers/blood , Congenital Disorders of Glycosylation/blood , Glycosylation , Humans , Protein Isoforms/metabolism
7.
J Virol ; 94(7)2020 03 17.
Article in English | MEDLINE | ID: mdl-31915280

ABSTRACT

Dengue virus (DENV) is a mosquito-borne flavivirus responsible for dengue disease, a major human health concern for which no specific therapies are available. Like other viruses, DENV relies heavily on the host cellular machinery for productive infection. In this study, we performed a genome-wide CRISPR-Cas9 screen using haploid HAP1 cells to identify host genes important for DENV infection. We identified DPM1 and -3, two subunits of the endoplasmic reticulum (ER) resident dolichol-phosphate mannose synthase (DPMS) complex, as host dependency factors for DENV and other related flaviviruses, such as Zika virus (ZIKV). The DPMS complex catalyzes the synthesis of dolichol-phosphate mannose (DPM), which serves as mannosyl donor in pathways leading to N-glycosylation, glycosylphosphatidylinositol (GPI) anchor biosynthesis, and C- or O-mannosylation of proteins in the ER lumen. Mutation in the DXD motif of DPM1, which is essential for its catalytic activity, abolished DPMS-mediated DENV infection. Similarly, genetic ablation of ALG3, a mannosyltransferase that transfers mannose to lipid-linked oligosaccharide (LLO), rendered cells poorly susceptible to DENV. We also established that in cells deficient for DPMS activity, viral RNA amplification is hampered and truncated oligosaccharides are transferred to the viral prM and E glycoproteins, affecting their proper folding. Overall, our study provides new insights into the host-dependent mechanisms of DENV infection and supports current therapeutic approaches using glycosylation inhibitors to treat DENV infection.IMPORTANCE Dengue disease, which is caused by dengue virus (DENV), has emerged as the most important mosquito-borne viral disease in humans and is a major global health concern. DENV encodes only few proteins and relies on the host cell machinery to accomplish its life cycle. The identification of the host factors important for DENV infection is needed to propose new targets for antiviral intervention. Using a genome-wide CRISPR-Cas9 screen, we identified DPM1 and -3, two subunits of the DPMS complex, as important host factors for the replication of DENV as well as other related viruses such as Zika virus. We established that DPMS complex plays dual roles during viral infection, both regulating viral RNA replication and promoting viral structural glycoprotein folding/stability. These results provide insights into the host molecules exploited by DENV and other flaviviruses to facilitate their life cycle.


Subject(s)
CRISPR-Cas Systems , Dengue Virus/physiology , Dengue/virology , Mannosyltransferases/metabolism , Animals , Chlorocebus aethiops , Endoplasmic Reticulum/metabolism , Fibroblasts/metabolism , Glycoproteins/metabolism , Glycosylation , Glycosylphosphatidylinositols/metabolism , HEK293 Cells , Humans , Mannose/chemistry , Oligosaccharides/chemistry , RNA, Guide, Kinetoplastida/metabolism , RNA, Viral/chemistry , Vero Cells , Virus Replication
8.
World Allergy Organ J ; 12(9): 100057, 2019 Sep.
Article in English | MEDLINE | ID: mdl-31641405

ABSTRACT

BACKGROUND: The natural history of allergic sensitization in childhood, and its impact on allergic disease development, needs to be clarified. This study aims to identify allergic sensitization and morbidity patterns during the first 8 years of life. METHODS: The study was conducted in the on-going population-based prospective Pollution and Asthma Risk: an Infant Study (PARIS) birth cohort. Sensitization profiles were identified by k-means clustering based upon allergen-specific IgE levels measured at 18 months and 8/9 years. Allergic morbidity profiles were identified by latent class analysis based on symptoms, symptom severity, treatments, and lifetime doctor-diagnoses of asthma, allergic rhinitis, and atopic dermatitis and on lower respiratory infections before 2 years. RESULTS: Five sensitization and 5 allergic morbidity patterns were established in 714 children. Children not sensitized or with isolated and low allergen-specific sensitization were grouped together (76.8%). A profile of early and transient sensitization to foods that increased the risk of asthma later in childhood was identified (4.9%). Children strongly sensitized (≥3.5 kUA/L) to house dust mite at 8/9 years (9.0%) had the highest risk of asthma and allergic rhinitis. Finally, timothy grass pollen at 8/9 years sensitization profile (5.3%) was related to respiratory allergic diseases, as was early onset and persistent sensitization profile (4.1%), this latter being also strongly associated with atopic dermatitis. CONCLUSIONS & CLINICAL RELEVANCE: We show that accurate assessment of the risk of allergic disease should rely on earliness and multiplicity of sensitization, involved allergens, and allergen-specific IgE levels, and not considering solely allergic sensitization as a dichotomous variable (allergen-specific IgE ≥0.35 kUA/L), as usually done. This is particularly striking for house dust mite. We are hopeful that, pending further confirmation in other populations, our findings will improve clinical practice as part of an approach to allergic disease prevention.

9.
J Thromb Haemost ; 17(11): 1798-1807, 2019 11.
Article in English | MEDLINE | ID: mdl-31271700

ABSTRACT

BACKGROUND: Congenital disorders of glycosylation are rare inherited diseases affecting many different proteins. The lack of glycosylation notably affects the hemostatic system and leads to deficiencies of both procoagulant and anticoagulant factors. OBJECTIVE: To assess the hemostatic balance in patients with multiple coagulation disorders by using a thrombin generation assay. METHOD: We performed conventional coagulation assays and a thrombin generation assay on samples from patients with congenital disorder of glycosylation. The thrombin generation assay was performed before and after activation of the protein C system by the addition of soluble thrombomodulin. RESULTS: A total of 35 patients were included: 71% and 57% had low antithrombin and factor XI levels, respectively. Protein C and protein S levels were abnormally low in 29% and 26% of the patients, respectively, whereas only 11% displayed low factor IX levels. Under baseline conditions, the thrombin generation assay revealed a significantly higher endogenous thrombin potential and thrombin peak in patients, relative to controls. After spiking with thrombomodulin, we observed impaired involvement of the protein C system. Hence, 54% of patients displayed a hypercoagulant phenotype in vitro. All the patients with a history of stroke-like episodes or thrombosis displayed this hypercoagulant phenotype. CONCLUSION: A thrombin generation assay revealed a hypercoagulant in vitro phenotype under baseline condition; this was accentuated by impaired involvement of the protein C system. This procoagulant phenotype may thus reflect the risk of severe vascular complications. Further research will have to determine whether the thrombin generation assay is predictive of vascular events.


Subject(s)
Blood Coagulation Disorders, Inherited/blood , Coagulation Protein Disorders/blood , Congenital Disorders of Glycosylation/blood , Thrombin/metabolism , Adolescent , Blood Coagulation/genetics , Blood Coagulation Disorders, Inherited/diagnosis , Blood Coagulation Disorders, Inherited/genetics , Child , Child, Preschool , Coagulation Protein Disorders/diagnosis , Coagulation Protein Disorders/genetics , Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/genetics , Female , Genetic Predisposition to Disease , Humans , Male , Paris , Phenotype , Retrospective Studies , Spain
10.
Hum Mutat ; 40(7): 938-951, 2019 07.
Article in English | MEDLINE | ID: mdl-31067009

ABSTRACT

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.


Subject(s)
Congenital Disorders of Glycosylation/genetics , Mannosyltransferases/genetics , Mutation , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/deficiency , Animals , COS Cells , Cells, Cultured , Child, Preschool , Chlorocebus aethiops , Female , Humans , Infant , Male , Open Reading Frames , Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase/genetics , Polymorphism, Single Nucleotide
11.
Toxicol Lett ; 305: 94-102, 2019 May 01.
Article in English | MEDLINE | ID: mdl-30716388

ABSTRACT

Exposure to airborne particulate matter (PM) has significant effects on human health mainly leading to cardio-respiratory diseases. However very few data are available regarding the impact of PM on the skin, so to better understand the impact of fine particle (PM0.3-2.5) on both inflammatory response and epidermal structure, we exposed a reconstructed human epidermis (RHE) to several doses of PM collected in Cotonou (Benin, West Africa). After 24 h of exposure, inflammatory response, histological observations, and gene expression related to oxidative stress, antioxidant defense and structural damages were determined. No PM-linked changes in tissue morphology or membrane integrity were observable. PM was however cytotoxic in a dose dependent manner. An inflammatory response appeared as shown by the increase in IL-1α and IL-8 cytokine productions. PM also induced oxidative stress, leading to an increase in 4-HNE immunostaining and to the up-regulation of HMOX1, MT1G and MT1E. Finally, PM had a negative impact on fundamental skin functions such as tissue anchorage, cell differentiation, cornification / skin desquamation and apoptosis. Our data show that airborne fine particles have an adverse effect on skin integrity, most probably leading to accelerated ageing.


Subject(s)
Air Pollutants/toxicity , Epidermis/drug effects , Keratinocytes/physiology , Particulate Matter/toxicity , Cell Culture Techniques , Cell Survival , Epidermis/metabolism , Gene Expression Regulation/drug effects , Humans , Keratinocytes/drug effects , Oxidative Stress , Particle Size , Toxicity Tests/methods
12.
J Inherit Metab Dis ; 42(1): 5-28, 2019 01.
Article in English | MEDLINE | ID: mdl-30740725

ABSTRACT

Phosphomannomutase 2 (PMM2-CDG) is the most common congenital disorder of N-glycosylation and is caused by a deficient PMM2 activity. The clinical presentation and the onset of PMM2-CDG vary among affected individuals ranging from a severe antenatal presentation with multisystem involvement to mild adulthood presentation limited to minor neurological involvement. Management of affected patients requires a multidisciplinary approach. In this article, a systematic review of the literature on PMM2-CDG was conducted by a group of international experts in different aspects of CDG. Our managment guidelines were initiated based on the available evidence-based data and experts' opinions. This guideline mainly addresses the clinical evaluation of each system/organ involved in PMM2-CDG, and the recommended management approach. It is the first systematic review of current practices in PMM2-CDG and the first guidelines aiming at establishing a practical approach to the recognition, diagnosis and management of PMM2-CDG patients.


Subject(s)
Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/drug therapy , Phosphotransferases (Phosphomutases)/deficiency , Follow-Up Studies , Glycosylation , Humans
13.
Pediatr Res ; 85(3): 384-389, 2019 02.
Article in English | MEDLINE | ID: mdl-30420707

ABSTRACT

BACKGROUND: Congenital disorders of glycosylation (CDG) includes ALG8 deficiency, a protein N-glycosylation defect with a broad clinical spectrum. If most of the 15 previously reported patients present an early-onset multisystem severe disease and early death, three patients including the cas princeps, present long-term survival and less severe symptoms. METHODS: In order to further characterize ALG8-CDG, two new ALG8 patients are described and mRNA analyses of the ALG8-CDG cas princeps were effected. RESULTS: One new patient exhibited a hepato-intestinal and neurological phenotype with two novel variants (c.91A > C p.Thr31Pro; c.139dup p.Thr47Asnfs*12). The other new patient, homozygous for a known variant (c.845C > T p.Ala282Val), presented a neurological phenotype with epilepsy, intellectual disability and retinis pigmentosa. The cas princeps ALG8-CDG patient was reported to have two heterozygous frameshift variants predicted to be without activity. We now described a novel ALG8 transcript variant in this patient and the 3D model of the putative encoded protein reveals no major difference with that of the normal ALG8 protein. CONCLUSION: The description of the two new ALG8 patients affirms that ALG8-CDG is a severe disease. In the cas princeps, as the originally described frameshift variants are degraded, the novel variant is promoted and could explain a milder phenotype.


Subject(s)
Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/genetics , Glucosyltransferases/genetics , Alternative Splicing , Emetine/pharmacology , Exons , Female , Frameshift Mutation , France , Genetic Variation , Glycosylation , Heterozygote , Homozygote , Humans , Infant , Male , Mutation, Missense , Phenotype , Retinitis Pigmentosa/genetics , Treatment Outcome
14.
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
15.
Eur J Hum Genet ; 27(3): 349-352, 2019 03.
Article in English | MEDLINE | ID: mdl-30552423

ABSTRACT

Next-generation sequencing (NGS) gene-panel-based analyses constitute diagnosis strategies which are adapted to the genetic heterogeneity within the field of myopathies, including more than 200 implicated genes to date. Nonetheless, important inter-laboratory diversity of gene panels exists at national and international levels, complicating the exchange of data and the visibility of the diagnostic offers available for referring neurologists. To address this issue, we here describe the initiative of the genetic diagnosis section of the French National Network for Rare Neuromuscular Diseases (Filière Nationale des Maladies Rares Neuromusculaires, FILNEMUS), which led to set up a consensual nationwide diagnostic strategy among the nine French genetic diagnosis laboratories using NGS for myopathies. The strategy is based on the determination of 13 clinical and/or histological entry-diagnosis groups, and consists for each group either in a successive NGS analysis of a "core gene list" followed in case of a negative result by the analysis of an "exhaustive gene list", or in the NGS analysis of a "unique exhaustive gene list".


Subject(s)
Consensus , Genetic Testing/standards , Neuromuscular Diseases/genetics , Practice Guidelines as Topic , Sequence Analysis, DNA/standards , France , Genetic Testing/methods , Humans , Neuromuscular Diseases/diagnosis , Sequence Analysis, DNA/methods , Societies, Medical
16.
Nat Commun ; 9(1): 3087, 2018 08 06.
Article in English | MEDLINE | ID: mdl-30082715

ABSTRACT

Skeletal dysplasia with multiple dislocations are severe disorders characterized by dislocations of large joints and short stature. The majority of them have been linked to pathogenic variants in genes encoding glycosyltransferases, sulfotransferases or epimerases required for glycosaminoglycan synthesis. Using exome sequencing, we identify homozygous mutations in SLC10A7 in six individuals with skeletal dysplasia with multiple dislocations and amelogenesis imperfecta. SLC10A7 encodes a 10-transmembrane-domain transporter located at the plasma membrane. Functional studies in vitro demonstrate that SLC10A7 mutations reduce SLC10A7 protein expression. We generate a Slc10a7-/- mouse model, which displays shortened long bones, growth plate disorganization and tooth enamel anomalies, recapitulating the human phenotype. Furthermore, we identify decreased heparan sulfate levels in Slc10a7-/- mouse cartilage and patient fibroblasts. Finally, we find an abnormal N-glycoprotein electrophoretic profile in patient blood samples. Together, our findings support the involvement of SLC10A7 in glycosaminoglycan synthesis and specifically in skeletal development.


Subject(s)
Amelogenesis Imperfecta/genetics , Bone Diseases, Developmental/genetics , Mutation , Organic Anion Transporters, Sodium-Dependent/genetics , Symporters/genetics , Animals , Body Weight , COS Cells , Child , Child, Preschool , Chlorocebus aethiops , Disease Models, Animal , Electrophoresis , Exome , Glycoproteins/chemistry , HEK293 Cells , Humans , Infant , Mice , Mice, Knockout , Osteochondrodysplasias/genetics
18.
Electrophoresis ; 39(24): 3123-3132, 2018 12.
Article in English | MEDLINE | ID: mdl-29869806

ABSTRACT

Congenital disorders of glycosylation (CDG) are rare autosomal genetic diseases affecting the glycosylation of proteins and lipids. Since CDG-related clinical symptoms are classically extremely variable and nonspecific, a combination of electrophoretic, mass spectrometric, and gene sequencing techniques is often mandatory for obtaining a definitive CDG diagnosis, as well as identifying causative gene mutations and deciphering the underlying biochemical mechanisms. Here, we illustrate the potential of integrating data from capillary electrophoresis of transferrin, two-dimensional electrophoresis of N- and O-glycoproteins, mass spectrometry analyses of total serum N-linked glycans and mucin core1 O-glycosylated apolipoprotein C-III for the determination of various culprit CDG gene mutations. "Step-by-step" diagnosis pathways of four particular and new CDG cases, including MGAT2-CDG, ATP6V0A2-CDG, SLC35A2-CDG, and SLC35A3-CDG, are described as illustrative examples.


Subject(s)
Congenital Disorders of Glycosylation , Electrophoresis/methods , Mass Spectrometry/methods , Sequence Analysis, DNA/methods , Adolescent , Child , Child, Preschool , Congenital Disorders of Glycosylation/blood , Congenital Disorders of Glycosylation/diagnosis , Female , Glycomics , Glycoproteins/blood , Glycoproteins/chemistry , Humans , Infant , Male , Polysaccharides/analysis , Polysaccharides/chemistry
19.
Mol Genet Metab ; 124(3): 228-235, 2018 07.
Article in English | MEDLINE | ID: mdl-29759592

ABSTRACT

Congenital disorders of glycosylation (CDG) linked to defects in Golgi apparatus homeostasis constitute an increasing part of these rare inherited diseases. Among them, COG-CDG, ATP6V0A2-CDG, TMEM199-CDG and CCDC115-CDG have been shown to disturb Golgi vesicular trafficking and/or lumen pH acidification. Here, we report 3 new unrelated cases of CCDC115-CDG with emphasis on diagnosis difficulties related to strong phenotypic similarities with mitochondriopathies, Niemann-Pick disease C and Wilson Disease. Indeed, while two individuals clinically presented with early and severe liver fibrosis and cirrhosis associated with neurological symptoms, the other one "only" showed isolated and late severe liver involvement. Biological results were similar to previously described patients, including hypercholesterolemia, elevated alkaline phosphatases and defects in copper metabolism. CDG screening and glycosylation study finally led to the molecular diagnosis of CCDC115-CDG. Besides pointing to the importance of CDG screening in patients with unexplained and severe liver disease, these reports expand the clinical and molecular phenotypes of CCDC115-CDG. The hepatic involvement is particularly addressed. Furthermore, hypothesis concerning the pathogenesis of the liver disease and of major biological abnormalities are proposed.


Subject(s)
Congenital Disorders of Glycosylation/complications , Golgi Apparatus/genetics , Liver Diseases/etiology , Mutation , Nerve Tissue Proteins/genetics , Adult , Congenital Disorders of Glycosylation/genetics , Congenital Disorders of Glycosylation/pathology , Female , Glycosylation , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , Humans , Infant, Newborn , Liver Diseases/pathology , Male , Prognosis , Young Adult
20.
Eur J Med Genet ; 61(11): 643-663, 2018 Nov.
Article in English | MEDLINE | ID: mdl-29079546

ABSTRACT

The survey summarizes in its first part the current status of knowledge on the Congenital Disorders of Glycosylation (CDG) with regard to their phenotypic spectrum, diagnostic and therapeutic strategies, and pathophysiology. It documents the clinical and basic research activities, and efforts to involve patients and their families. In the second part, it tries to look into the future of CDG. More specific biomarkers are needed for fast CDG diagnosis and treatment monitoring. Whole genome sequencing will play an increasingly important role in the molecular diagnosis of unsolved CDG. Epigenetic defects are expected to join the rapidly expanding genetic and allelic heterogeneity of the CDG family. Novel treatments are urgently needed particularly for PMM2-CDG, the most prevalent CDG. Patient services such as apps should be developed e.g. to document the natural history and monitor treatment. Networking (EURO-CDG, the European Reference Networks (MetabERN)) is an efficient tool to disseminate knowledge and boost collaboration at all levels. The final goal is of course to improve the quality of life of the patients and their families.


Subject(s)
Congenital Disorders of Glycosylation/epidemiology , Congenital Disorders of Glycosylation/genetics , Phosphotransferases (Phosphomutases)/genetics , Congenital Disorders of Glycosylation/diagnosis , Congenital Disorders of Glycosylation/pathology , Glycosylation , Humans , Mutation/genetics , Quality of Life , Surveys and Questionnaires
SELECTION OF CITATIONS
SEARCH DETAIL
...