1,137 results on '"Jaak Jaeken"'
Search Results
2. A Community-Based Participatory Framework to Co-Develop Patient Education Materials (PEMs) for Rare Diseases: A Model Transferable across Diseases
- Author
-
Marta Falcão, Mariateresa Allocca, Ana Sofia Rodrigues, Pedro Granjo, Rita Francisco, Carlota Pascoal, Maria Grazia Rossi, Dorinda Marques-da-Silva, Salvador C. M. Magrinho, Jaak Jaeken, Larisa Aragon Castro, Cláudia de Freitas, Paula A. Videira, Luísa de Andrés-Aguayo, and Vanessa dos Reis Ferreira
- Subjects
plain-language ,Community-based participatory research ,congenital disorders of glycosylation (CDG) ,Health, Toxicology and Mutagenesis ,patient education material (PEM) ,Public Health, Environmental and Occupational Health ,public and patient involvement (PPI) ,Community Participation ,Health literacy ,rare diseases ,Plain-language ,Patient education material (PEM) ,patient empowerment ,Rare diseases ,Health Literacy ,People-centric ,Congenital Disorders of Glycosylation ,Patient Education as Topic ,Congenital disorders of glycosylation (CDG) ,people-centric ,Humans ,Patient empowerment ,community-based participatory research ,health literacy ,Public and patient involvement (PPI) - Abstract
At least 50% of chronic disease patients don't follow their care plans, leading to lower health outcomes and higher medical costs. Providing Patient Education Materials (PEMs) to individuals living with a disease can help to overcome these problems. PEMs are especially beneficial for people suffering from multisystemic and underrecognized diseases, such as rare diseases. Congenital disorders of glycosylation (CDG) are ultra-rare diseases, where a need was identified for PEMs in plain language that can clearly explain complex information. Community involvement in the design of PEMs is extremely important for diseases whose needs are underserved, such as rare diseases; however, attempts to involve lay and professional stakeholders are lacking. This paper presents a community-based participatory framework to co-create PEMs for CDG, that is transferable to other diseases. A literature review and questionnaire were performed, and only four articles describing the development of PEMS for rare diseases have been found, which demonstrates a lack of standardized approaches. The framework and PEMs were co-developed with CDG families and will be crucial in increasing health literacy and empowering families. We will close a gap in the creation of PEMs for CDG by delivering these resources in lay language in several languages. ispartof: Int J Environ Res Public Health vol:20 issue:2 pages:968- ispartof: location:Switzerland status: Published online
- Published
- 2023
3. Epidemiology of congenital disorders of glycosylation (CDG)—overview and perspectives
- Author
-
Ana Piedade, Rita Francisco, Jaak Jaeken, Peymaneh Sarkhail, Sandra Brasil, Carlos R. Ferreira, Tatiana Rijoff, Carlota Pascoal, Alexandre Gil, Ana Beatriz Lourenço, Marta Abreu, Mafalda Gomes, Paula A. Videira, and Vanessa dos Reis Ferreira
- Abstract
Background and aim Congenital disorders of glycosylation (CDG) are a large heterogeneous group of about 170 rare inherited metabolic disorders due to defective protein and lipid glycosylation. This study aimed to assemble and summarise available data on the epidemiology of CDG. Methods A set of keywords related to epidemiology and CDG was defined. The keywords were combined through a custom Python script, search through the MEDLINE database, using PubMed as the search engine. The script retrieved the correspondent MEDLINE data from each article, and the relevant information was exported. Next, inclusion and exclusion criteria were set and applied during the selection phase. Finally, epidemiology-related information was extracted and compiled. Results One hundred sixty-five papers on CDG epidemiology were included in this literature review. Most of them reported on the frequency of symptoms in CDG patients followed in cohort studies, on pathogenic variant allelic frequency, and on the prevalence of the disorder in populations. According to this review, the most reported CDG was phosphomannomutase-2 deficiency (PMM2-CDG) followed in descending order by FKTN-CDG, EXT1/EXT2-CDG, ALG6-CDG, and PIGA-CDG. Conclusions We provide an overview on epidemiological data regarding 93 CDG by compiling information from the literature. Generating epidemiological data on CDG is important to appropriately target resources for CDG research and drug development and to support public health decision-making.
- Published
- 2022
4. Active site variants in STT3A cause a dominant type I congenital disorder of glycosylation with neuromusculoskeletal findings
- Author
-
Rita Barone, Filippo Vairo, Bobby G. Ng, Jaak Jaeken, Gert Matthijs, James Pitt, Thierry Dupré, Lyndon Gallacher, Liesbeth Keldermans, Helen Michelakakis, Marina Ventouratou, Susan M. White, Sze Chern Lim, Melissa Baerenfaenger, Mirian C. H. Janssen, Angel Ashikov, Karin Huijben, Sandrine Vuillaumier-Barrot, Diana Ballhausen, Daisy Rymen, Agustí Rodríguez-Palmero, Blai Morales-Romero, Antonia Ribes, Peter Witters, Heidi Peters, Erika Souche, Eva Morava, Agata Fiumara, Pascale de Lonlay, Matthew P. Wilson, Dirk Lefeber, Wasantha Ranatunga, Alejandro Garanto, Hudson H. Freeze, Christian Thiel, BioAnalytical Chemistry, and AIMMS
- Subjects
Male ,Mutant ,congenital disorders of glycosylation ,chemistry.chemical_compound ,0302 clinical medicine ,Catalytic Domain ,Missense mutation ,Musculoskeletal Diseases ,Genetics (clinical) ,Genes, Dominant ,chemistry.chemical_classification ,Genetics ,0303 health sciences ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Middle Aged ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Pedigree ,Oligosaccharyltransferase complex ,Child, Preschool ,glycosylation ,Female ,Adult ,Heterozygote ,Glycosylation ,Adolescent ,Protein subunit ,Biology ,Article ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,oligosaccharyltransferase complex ,medicine ,Humans ,dominant inheritance ,Amino Acid Sequence ,030304 developmental biology ,Sequence Homology, Amino Acid ,Oligosaccharyltransferase ,Membrane Proteins ,medicine.disease ,chemistry ,Hexosyltransferases ,Nervous System Diseases ,Glycoprotein ,Congenital disorder of glycosylation ,030217 neurology & neurosurgery - Abstract
Congenital disorders of glycosylation (CDGs) form a group of rare diseases characterized by hypoglycosylation. We here report the identification of 16 individuals from nine families who have either inherited or de novo heterozygous missense variants in STT3A, leading to an autosomal-dominant CDG. STT3A encodes the catalytic subunit of the STT3A-containing oligosaccharyltransferase (OST) complex, essential for protein N-glycosylation. Affected individuals presented with variable skeletal anomalies, short stature, macrocephaly, and dysmorphic features; half had intellectual disability. Additional features included increased muscle tone and muscle cramps. Modeling of the variants in the 3D structure of the OST complex indicated that all variants are located in the catalytic site of STT3A, suggesting a direct mechanistic link to the transfer of oligosaccharides onto nascent glycoproteins. Indeed, expression of STT3A at mRNA and steady-state protein level in fibroblasts was normal, while glycosylation was abnormal. In S. cerevisiae, expression of STT3 containing variants homologous to those in affected individuals induced defective glycosylation of carboxypeptidase Y in a wild-type yeast strain and expression of the same mutants in the STT3 hypomorphic stt3-7 yeast strain worsened the already observed glycosylation defect. These data support a dominant pathomechanism underlying the glycosylation defect. Recessive mutations in STT3A have previously been described to lead to a CDG. We present here a dominant form of STT3A-CDG that, because of the presence of abnormal transferrin glycoforms, is unusual among dominant type I CDGs. ispartof: AMERICAN JOURNAL OF HUMAN GENETICS vol:108 issue:11 pages:2130-2144 ispartof: location:United States status: published
- Published
- 2021
5. A Participatory Framework for Plain Language Clinical Management Guideline Development
- Author
-
Rita Francisco, Susana Alves, Catarina Gomes, Pedro Granjo, Carlota Pascoal, Sandra Brasil, Alice Neves, Inês Santos, Andrea Miller, Donna Krasnewich, Eva Morava, Christina Lam, Jaak Jaeken, Paula A. Videira, Vanessa dos Reis Ferreira, UCIBIO - Applied Molecular Biosciences Unit, and DCV - Departamento de Ciências da Vida
- Subjects
Science & Technology ,DISORDERS ,clinical management guidelines ,congenital disorders of glycosylation (CDG) ,Health, Toxicology and Mutagenesis ,Public Health, Environmental and Occupational Health ,rare diseases ,Environmental Sciences & Ecology ,DIAGNOSIS ,Pollution ,plain language ,people-centric ,PMM2-CDG ,participatory medicine ,health literacy ,Congenital Disorders of Glycosylation ,SDG 3 - Good Health and Well-being ,Quality of Life ,Humans ,Life Sciences & Biomedicine ,Environmental Sciences ,Public, Environmental & Occupational Health ,Language - Abstract
BACKGROUND: Clinical management guidelines (CMGs) are decision support tools for patient care used by professionals, patients, and family caregivers. Since clinical experts develop numerous CMGs, their technical language hinders comprehension and access by nonmedical stakeholders. Additionally, the views of affected individuals and their families are often not incorporated into treatment guidelines. We developed an adequate methodology for addressing the needs and preferences of family and professional stakeholders regarding CMGs, a recently developed protocol for managing congenital disorders of glycosylation (CDG), a family of rare metabolic diseases. We used the CDG community and phosphomannomutase 2 (PMM2)-CDG CMGs as a pilot to test and implement our methodology. RESULTS: We listened to 89 PMM2-CDG families and 35 professional stakeholders and quantified their CMG-related needs and preferences through an electronic questionnaire. Most families and professionals rated CMGs as relevant (86.5% and 94.3%, respectively), and valuable (84.3% and 94.3%, respectively) in CDG management. The most identified challenges were the lack of CMG awareness (50.6% of families) and the lack of plain language CMG (39.3% of professionals). Concordantly, among families, the most suggested solution was involving them in CMG development (55.1%), while professionals proposed adapting CMGs to include plain language (71.4%). Based on these results, a participatory framework built upon health literacy principles was created to improve CMG comprehension and accessibility. The outputs are six complementary CMG-related resources differentially adapted to the CDG community's needs and preferences, with a plain language PMM2-CDG CMG as the primary outcome. Additionally, the participants established a distribution plan to ensure wider access to all resources. CONCLUSIONS: This empowering, people-centric methodology accelerates CMG development and accessibility to all stakeholders, ultimately improving the quality of life of individuals living with a specific condition and raising the possibility of application to other clinical guidelines. ispartof: INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH vol:19 issue:20 ispartof: location:Switzerland status: published
- Published
- 2022
6. Stakeholders’ views on drug development: the congenital disorders of glycosylation community perspective
- Author
-
Maria Monticelli, Rita Francisco, Sandra Brasil, Dorinda Marques-da-Silva, Tatiana Rijoff, Carlota Pascoal, Jaak Jaeken, Paula A. Videira, Vanessa dos Reis Ferreira, Monticelli, M., Francisco, R., Brasil, S., Marques-da-Silva, D., Rijoff, T., Pascoal, C., Jaeken, J., Videira, P. A., and dos Reis Ferreira, V.
- Subjects
Genetics & Heredity ,INVOLVEMENT ,Science & Technology ,Glycosylation ,Drug development ,General Medicine ,Research & Experimental Medicine ,Congenital disorder(s) of glycosylation (CDG) ,Electronic (e-)survey ,Drug development, People-centricity, Congenital disorder(s) of glycosylation, ELectronic (e-)survey, Patient-reported outcome measures ,Congenital Disorders of Glycosylation ,Medicine, Research & Experimental ,Artificial Intelligence ,Patient-reported outcome measures ,Humans ,People-centricity ,CDG ,Family ,Pharmacology (medical) ,Patient Participation ,INTERNET ,Life Sciences & Biomedicine ,Genetics (clinical) - Abstract
Background Congenital disorders of glycosylation (CDG) are a large family of rare genetic diseases for which therapies are virtually nonexistent. However, CDG therapeutic research has been expanding, thanks to the continuous efforts of the CDG medical/scientific and patient communities. Hence, CDG drug development is a popular research topic. The main aim of this study was to understand current and steer future CDG drug development and approval by collecting and analysing the views and experiences of the CDG community, encompassing professionals and families. An electronic (e-)survey was developed and distributed to achieve this goal. Results A total of 128 respondents (46 CDG professionals and 82 family members), mainly from Europe and the USA, participated in this study. Most professionals (95.0%) were relatively familiar with drug development and approval processes, while CDG families revealed low familiarity levels, with 8.5% admitting to never having heard about drug development. However, both stakeholder groups agreed that patients and families make significant contributions to drug development and approval. Regarding their perceptions of and experiences with specific drug development and approval tools, namely biobanks, disease models, patient registries, natural history studies (NHS) and clinical trials (CT), the CDG community stakeholders described low use and participation, as well as variable familiarity. Additionally, CDG professionals and families shared conflicting views about CT patient engagement and related information sharing. Families reported lower levels of involvement in CT design (25.0% declared ever being involved) and information (60.0% stated having been informed) compared to professionals (60.0% and 85.7%, respectively). These contrasting perceptions were further extended to their insights and experiences with patient-centric research. Finally, the CDG community (67.4% of professionals and 54.0% of families) reported a positive vision of artificial intelligence (AI) as a drug development tool. Nevertheless, despite the high AI awareness among CDG families (76.8%), professionals described limited AI use in their research (23.9%). Conclusions This community-centric study sheds new light on CDG drug development and approval. It identifies educational, communication and research gaps and opportunities for CDG professionals and families that could improve and accelerate CDG therapy development.
- Published
- 2022
7. De novo loss-of-function variants in X-linked MED12 are associated with Hardikar syndrome in females
- Author
-
Eugênia Ribeiro Valadares, Kaitlyn M Shen, Sérgio D.J. Pena, Alanna Strong, Dong Li, Natália D. Linhares, David Cassiman, Elaine H. Zackai, Deindl Philipp, Penny Chow, Arupa Ganguly, Jaak Jaeken, Samantha A. Schrier Vergano, Maria Van Dyck, Tatjana Bierhals, Tiancheng Wang, Elizabeth J. Bhoj, Hakon Hakonarson, Anne Hing, and Tasja Scholz
- Subjects
0301 basic medicine ,Genetics ,business.industry ,media_common.quotation_subject ,Nonsense ,030105 genetics & heredity ,medicine.disease ,Frameshift mutation ,MED12 ,03 medical and health sciences ,030104 developmental biology ,Intestinal malrotation ,Intellectual disability ,medicine ,Missense mutation ,business ,Genetics (clinical) ,Exome sequencing ,Loss function ,media_common - Abstract
Purpose Hardikar syndrome (MIM 612726) is a rare multiple congenital anomaly syndrome characterized by facial clefting, pigmentary retinopathy, biliary anomalies, and intestinal malrotation, but with preserved cognition. Only four patients have been reported previously, and none had a molecular diagnosis. Our objective was to identify the genetic basis of Hardikar syndrome (HS) and expand the phenotypic spectrum of this disorder. Methods We performed exome sequencing on two previously reported and five unpublished female patients with a clinical diagnosis of HS. X-chromosome inactivation (XCI) studies were also performed. Results We report clinical features of HS with previously undescribed phenotypes, including a fatal unprovoked intracranial hemorrhage at age 21. We additionally report the discovery of de novo pathogenic nonsense and frameshift variants in MED12 in these seven individuals and evidence of extremely skewed XCI in all patients with informative testing. Conclusion Pathogenic missense variants in the X-chromosome gene MED12 have previously been associated with Opitz-Kaveggia syndrome, Lujan syndrome, Ohdo syndrome, and nonsyndromic intellectual disability, primarily in males. We propose a fifth, female-specific phenotype for MED12, and suggest that nonsense and frameshift loss-of-function MED12 variants in females cause HS. This expands the MED12-associated phenotype in females beyond intellectual disability.
- Published
- 2021
8. Congenital Disorders of Glycosylation in Portugal—Two Decades of Experience
- Author
-
Ana Maria Fortuna, Isaura Ribeiro, Gert Matthijs, Francisco Ferraz Laranjeira, Jaak Jaeken, Dulce Quelhas, Luísa Azevedo, Ana Medeira, Helena Cabral Fernandes, Ana C. Ferreira, Sílvia Sequeira, A.F. Oliveira, Paula Garcia, Carla Mendonça, Valerie Race, Liesbeth Keldermans, Anabela Bandeira, Elisa Leão Teles, Esmeralda Rodrigues, Erica Souche, Patrícia Janeiro, Ana Maria Minarelli Gaspar, Luísa Diogo, and Esmeralda Martins
- Subjects
Male ,Time Factors ,Adolescent ,HDE MTB ,congenital disorder(s) of glycosylation ,PMM2 genotype ,Cohort Studies ,Young Adult ,03 medical and health sciences ,symbols.namesake ,Congenital Disorders of Glycosylation ,0302 clinical medicine ,030225 pediatrics ,PGM1 ,Humans ,Medicine ,030212 general & internal medicine ,Allele ,Child ,Exome sequencing ,Genetics ,Sanger sequencing ,Massive parallel sequencing ,Portugal ,business.industry ,Transferrin ,Infant ,DPAGT1 ,Phenotype ,Child, Preschool ,Pediatrics, Perinatology and Child Health ,Cohort ,Congenital disorder/glycosylation ,symbols ,Female ,CDG ,business ,Phosphomannomutase - Abstract
OBJECTIVE: To describe the clinical, biochemical, and genetic features of both new and previously reported patients with congenital disorders of glycosylation (CDGs) diagnosed in Portugal over the last 20 years. STUDY DESIGN: The cohort includes patients with an unexplained multisystem or single organ involvement, with or without psychomotor disability. Serum sialotransferrin isoforms and, whenever necessary, apolipoprotein CIII isoforms and glycan structures were analyzed. Additional studies included measurement of phosphomannomutase (PMM) activity and analysis of lipid-linked oligosaccharides in fibroblasts. Sanger sequencing and massive parallel sequencing were used to identify causal variants or the affected gene, respectively. RESULTS: Sixty-three individuals were diagnosed covering 14 distinct CDGs; 43 patients diagnosed postnatally revealed a type 1, 14 a type 2, and 2 a normal pattern on serum transferrin isoelectrofocusing. The latter patients were identified by whole exome sequencing. Nine of them presented also a hypoglycosylation pattern on apolipoprotein CIII isoelectrofocusing, pointing to an associated O-glycosylation defect. Most of the patients (62%) are PMM2-CDG and the remaining carry pathogenic variants in ALG1, ATP6AP1, ATP6AP2, ATP6V0A2, CCDC115, COG1, COG4, DPAGT1, MAN1B1, SLC35A2, SRD5A3, RFT1, or PGM1. CONCLUSIONS: Portuguese patients with CDGs are presented in this report, some of them showing unique clinical phenotypes. Among the 14 genes mutated in Portuguese individuals, 8 are shared with a previously reported Spanish cohort. However, regarding the mutational spectrum of PMM2-CDG, the most frequent CDG, a striking similarity between the 2 populations was found, as only 1 mutated allele found in the Portuguese group has not been reported in Spain. ispartof: JOURNAL OF PEDIATRICS vol:231 pages:148-156 ispartof: location:United States status: published
- Published
- 2021
9. Genetic modifiers in glycosylation pathways: Is there a link between <scp>PMM2</scp> and <scp>PGM1</scp> ?
- Author
-
Dulce Quelhas, Jaak Jaeken, and Luisa Azevedo
- Subjects
Genetics ,Genetics (clinical) - Published
- 2022
10. The road to successful people-centric research in rare diseases: the web-based case study of the Immunology and Congenital Disorders of Glycosylation questionnaire (ImmunoCDGQ)
- Author
-
Rita Francisco, Sandra Brasil, Carlota Pascoal, Jaak Jaeken, Merell Liddle, Paula A. Videira, and Vanessa dos Reis Ferreira
- Subjects
Congenital Disorders of Glycosylation (CDG) ,Patient engagement ,Electronic (e-) questionnaire ,General Medicine ,Rare diseases ,Social media ,Rare Diseases ,Congenital Disorders of Glycosylation ,Patient recruitment ,Surveys and Questionnaires ,Quality of Life ,Humans ,Patient empowerment ,People-centricity ,Pharmacology (medical) ,Social Media ,Genetics (clinical) ,Web-based platforms - Abstract
BackgroundCongenital Disorders of Glycosylation (CDG) are a complex family of rare metabolic diseases. Robust clinical data collection faces many hurdles, preventing full CDG biological and clinical comprehension. Web-based platforms offer privileged opportunities for biomedical data gathering, and participant recruitment, particularly in rare diseases. The immunology and CDG electronic (e-) questionnaire (ImmunoCDGQ) explores this paradigm, proposing a people-centric framework to advance health research and participant empowerment.ObjectiveThe objectives of this study were to: (1) Describe and characterize the ImmunoCDGQ development, engagement, recruitment, participation, and result dissemination strategies; (2) To critically compare this framework with published literature and making recommendations.MethodsAn international, multistakeholder people-centric approach was initiated to develop and distribute the ImmunoCDGQ, a multi-lingual e-questionnaire able to collect immune-related data directly from patients and family caregivers. An adapted version was produced and distributed among the general “healthy” population (ImmunoHealthyQ), serving as the control group. Literature screening was performed to identify and analyze comparable studies.ResultsThe ImmunoCDGQ attained high participation and inclusion rates (94.6%, 209 out of 221). Comparatively to the control, CDG participants also showed higher and more variable questionnaire completion times as well as increased English version representativeness. Additionally, 20% of the CDG group (42 out of 209) chose not to complete the entire questionnaire in one go. Conditional logic structuring guided participant data provision and accurate data analysis assignment. Multi-channel recruitment created sustained engagement with Facebook emerging as the most followed social media outlet. Still, most included ImmunoCDGQ questionnaires (50.7%, 106 out of 209) were submitted within the first month of the project’s launch. Literature search and analysis showed that most e-questionnaire-based studies in rare diseases are author-built (56.8%, 25 out of 44), simultaneously addressing medical and health-related quality of life (HRQoL) and/or information needs (79.5%, 35 out of 44). Also, over 68% of the studies adopt multi-platform recruitment (30 out of 44) actively supported by patient organizations (52.3%, 23 out of 44).ConclusionsThe ImmunoCDGQ, its methodology and the CDG Community served as models for health research, hence paving a successful and reproducible road to people-centricity in biomedical research.
- Published
- 2022
11. MAN1B1-CDG: novel patients and novel variant
- Author
-
Liesbeth Keldermans, Gert Matthijs, Asburce Olgac, Çiğdem Seher Kasapkara, Jaak Jaeken, and Mustafa Kilic
- Subjects
medicine.medical_specialty ,Glycosylation ,Endocrinology, Diabetes and Metabolism ,030209 endocrinology & metabolism ,Genetic analysis ,03 medical and health sciences ,chemistry.chemical_compound ,symbols.namesake ,0302 clinical medicine ,Endocrinology ,030225 pediatrics ,Internal medicine ,Medicine ,Truncal obesity ,chemistry.chemical_classification ,business.industry ,Endoplasmic reticulum ,Golgi apparatus ,medicine.disease ,Hypotonia ,chemistry ,Transferrin ,Pediatrics, Perinatology and Child Health ,symbols ,Elevated transaminases ,medicine.symptom ,business - Abstract
Objectives Congenital disorders of glycosylation (CDGs) are a group of genetic disorders due to hypoglycosylation of proteins and lipids. A type I pattern is associated with defects in glycan assembly and transfer (CDG-I; cytosol; and endoplasmic reticulum defects), a type II pattern is seen in processing defects of the Golgi apparatus. MAN1B1-CDG is an autosomal recessive CDG-II due to mutations in the α 1,2-mannosidase gene (MAN1B1), mainly characterized by psychomotor disability, facial dysmorphism, truncal obesity, and hypotonia. Case presentation Three patients (two males and one female), with MAN1B1-CDG who had elevated transaminase levels are presented. All patients had presented due to dysmorphic and neurological findings and hypertransaminasemia was remarkable. A type 2 pattern was found on serum transferrin isoelectrofocusing analysis of the presented cases. MAN1B1-CDG was confirmed by genetic analysis. Conclusions Although the cause of the increased serum transaminase levels in the present patients is not clear, no evidence for an infection or underlying liver pathology could be identified. In order to know if this is a consistent feature, we suggest measuring serum transaminase levels regularly in MAN1B1-CDG patients.
- Published
- 2021
12. HILIC-UPLC-MS for high throughput and isomeric N-glycan separation and characterization in Congenital Disorders Glycosylation and human diseases
- Author
-
Agata Fiumara, Domenico Garozzo, Luisa Sturiale, Rita Barone, Angela Messina, Angelo Palmigiano, Francesca Esposito, Jaak Jaeken, and Andrea Bordugo
- Subjects
Spectrometry, Mass, Electrospray Ionization ,Glycan ,Glycosylation ,High throughput N-glycomics ,Electrospray ionization ,Mass spectrometry ,Biochemistry ,03 medical and health sciences ,chemistry.chemical_compound ,Congenital Disorders of Glycosylation ,Isomerism ,Polysaccharides ,Liquid chromatography–mass spectrometry ,Mannosidases ,Humans ,HILIC ,Derivatization ,Molecular Biology ,Chromatography, High Pressure Liquid ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,Chemistry ,Hydrophilic interaction chromatography ,Oligomannose type N-glycans ,030302 biochemistry & molecular biology ,Membrane Proteins ,alpha-Glucosidases ,Cell Biology ,LC-MS ,carbohydrates (lipids) ,HILIC . High throughput N-glycomics . CDG . LC-MS . Oligomannose type N-glycans ,biology.protein ,CDG ,Glycoprotein ,Blood Chemical Analysis - Abstract
N-glycan analyses may serve uncovering disease-associated biomarkers, as well as for profiling distinctive changes supporting diagnosis of genetic disorders of glycan biosynthesis named congenital disorders of glycosylation (CDG). Strategies based on liquid chromatography (LC) preferentially coupled to electrospray ionization (ESI) - mass spectrometry (MS) have emerged as powerful analytical methods for N-glycan identification and characterization. To enhance detection sensitivity, glycans are commonly labelled with a functional tag prior to LC-MS analysis. Since most derivatization techniques are notoriously time-consuming, some commercial analytical kits have been developed to speed up N-deglycosylation and N-glycan labelling of glycoproteins of pharmaceutical and biological interest such as monoclonal antibodies (mAbs). We exploited the analytical capabilities of RapiFluor-MS (RFMS) to perform, by a slightly modified protocol, a detailed N-glycan characterization of total serum and single serum glycoproteins from specific patients with CDG (MAN1B1-CDG, ALG12-CDG, MOGS-CDG, TMEM199-CDG). This strategy, accomplished by Hydrophilic Interaction Chromatography (HILIC)-UPLC-ESI-MS separation of the RFMS derivatized N-glycans, allowed us to uncover structural details of patients serum released N-glycans, thus extending the current knowledge on glycan profiles in these individual glycosylation diseases. The applied methodology enabled to differentiate in some cases either structural isomers and isomers differing in the linkage type. All the here reported applications demonstrated that RFMS method, coupled to HILIC-UPLC-ESI-MS, represents a sensitive high throughput approach for serum N-glycome analysis and a valuable option for glycan detection and separation particularly for isomeric species.
- Published
- 2020
13. Biallelic MADD variants cause a phenotypic spectrum ranging from developmental delay to a multisystem disorder
- Author
-
Katherine Wesseling Perry, Archana Raja, Emilie D. Douine, Xue Zhong Liu, Brent L. Fogel, Stan F. Nelson, Kenneth R. Maravilla, Eva H. Baker, Dave Viskochil, Kerstin Kutsche, Jordan H. Whitlock, Susan L. Samson, Christine M. Eng, Chloe M. Reuter, Suman Jayadev, David R. Adams, Sihoun Hahn, Rebecca C. Spillmann, Margaret Adam, Heather C Mefford, John C. Carey, Ehsan Ghayoor Karimiani, Donna M. Krasnewich, David Goldstein, Susan A. Korrick, Guoyun Yu, Tomas Honzik, Henry Houlden, Andrea L. Gropman, David A. Sweetser, Anna Bican, Carlos A. Bacino, Liliana Fernandez, Gabrielle Brown, Justin Alvey, Hane Lee, Emanuele G. Coci, Hongzheng Dai, Mario Saporta, Laurel A. Cobban, John F. Bohnsack, Stephanie Fox, Heidi Cope, Tyra Estwick, Lorraine Potocki, Nichole Hayes, Elizabeth A. Burke, Rizwan Hamid, Aaron R. Quinlan, Kelly Hassey, Lindsay C. Burrage, Jane Juusola, Adeline Vanderver, Malik Alawi, Teri A. Manolio, Maja Hempel, Esther M. Maier, Jennifer Kennedy, Bruce D. Gelb, Martha Horike-Pyne, Amarilis Sanchez-Valle, Euan A. Ashley, Surendra Dasari, Elizabeth Blue, Eva Morava-Kozicz, Natalie Rosenwasser, Alan H. Beggs, Bryn D. Webb, Isaac S. Kohane, Kelly Schoch, C. Christopher Lau, Nicole M. Walley, Laura M. Amendola, Genecee Renteria, Catherine H. Sillari, Jonathan A. Bernstein, Pinar Bayrak-Toydemir, R. Frank Kooy, Mariko Nakano-Okuno, Manuela Siekmeyer, Marije E. C. Meuwissen, Stephanie Bivona, Mark Wener, Precilla D'Souza, Olveen Carrasquillo, Paolo Moretti, Diane B. Zastrow, David J. Eckstein, Janet S. Sinsheimer, Kathy Sisco, Holly K. Tabor, William E. Byrd, Esteban C. Dell'Angelica, Rosario Isasi, Jacinda B. Sampson, Carsten Bonnenmann, J. Lawrence Merritt, Joan M. Stoler, Richard L. Maas, Paul G. Fisher, Jeanette C. Papp, Kimberly LeBlanc, Lilianna Solnica-Krezel, Mustafa Tekin, Mathias Woidy, Andrew B. Crouse, Katleen Ballon, David Murphy, Matthew T. Wheeler, Joseph Loscalzo, Ellen Macnamara, Cecelia P. Tamburro, Lefkothea P. Karaviti, Chunli Zhao, Ingrid A. Holm, Pankaj B. Agrawal, Alana L. Grajewski, Stephen C. Pak, Ian R. Lanza, Mohammad Doosti, Jennifer E. Posey, Rebecca Signer, Katie Golden-Grant, Christopher A. Walsh, Alica M. Goldman, Jyoti G. Dayal, Queenie K.-G. Tan, Martin G. Martin, Joy D. Cogan, Kevin S. Smith, Deborah A. Nickerson, Elisabeth McGee, Laure Fresard, Rena A. Godfrey, Sharyn A. Lincoln, Kathleen E. Sullivan, Mariska Davids, Melissa A. Walker, Prashant Sharma, Maria Iascone, Neil H. Parker, Carlos Ferreira, Elizabeth L. Fieg, Edwin K. Silverman, Michael L. Cunningham, Pengfei Liu, Edward M. Behrens, Sandra K. Loo, David R. Murdock, F. Sessions Cole, C. Ron Scott, Dan Doherty, Elly Brokamp, John H. Newman, Alden Y. Huang, Laura A. Pace, Avi Nath, Jimmy Bennet, Georg Christoph Korenke, Alyssa A. Tran, Gabriel F. Batzli, Jimann Shin, Matthew A. Deardorff, Naghmeh Dorrani, Diane Beysen, Irma Gutierrez, Stanislav Kmoch, Majid Alfadhel, Fred F. Telischi, Jennifer A. Sullivan, William A. Gahl, María Palomares-Bralo, Gerard T. Berry, Colleen E. McCormack, Lance H. Rodan, Reza Maroofian, Lenka Nosková, Judy Schaechter, Lynne A. Wolfe, Deborah Krakow, Daryl A. Scott, Tara Wenger, Jason Hom, Dustin Baldridge, Lynette Rives, Lee-kai Wang, Dawn L. Earl, Ralph L. Sacco, Fernando Santos-Simarro, Irman Forghani, Fuki M. Hisama, Terra R. Coakley, Hsiao-Tuan Chao, Jeremy D. Woods, Emily G. Kelley, Jean M. Johnston, Neil A. Hanchard, Amy K. Robertson, Matt Velinder, Byron L. Lam, Wendy H. Raskind, William J. Craigen, Stephan Züchner, Guney Bademci, Julian A. Martinez-Agosto, Mary Koziura, Beth A. Martin, Angela Sun, John A. Phillips, Seema R. Lalani, Daniela Buhas, Emily Solem, Gary D. Clark, Gill Bejerano, Ingo Kurth, Deborah Barbouth, Tiina K. Urv, Fanny Kortüm, Ian A. Glass, Ta Chen Peter Chang, Yong Huang, Roy C. Levitt, Paola Francesca Ajmone, Brenna Boyd, René Santer, Tim Schedl, David D. Draper, Ghayda M. Mirzaa, Aroa Rodríguez Alonso, Stephanie Wallace, Colleen E. Wahl, Calum A. MacRae, Gail P. Jarvik, Jacob L. McCauley, Jill A. Rosenfeld, Ronit Marom, Monte Westerfield, Matthew Might, Poupak Javaher-Haghighi, Brendan C. Lanpher, Devon Bonner, Cynthia J. Tifft, Cecilia Esteves, May Christine V. Malicdan, Jim Bale, Fariha Jamal, Nicola Longo, Christina G.S. Palmer, Lisa Emrick, Peter H. Byers, Vandana Shashi, Tiphanie P. Vogel, Richard A. Lewis, Jijun Wan, Barbara N. Pusey, Maria T. Acosta, Jaak Jaeken, Allyn McConkie-Rosell, Shirley Sutton, John Yang, Lorenzo D. Botto, Hilde Peeters, Rong Mao, Catherine Groden, Brendan Lee, Marta M. Majcherska, Rami Abou Jamra, Ashok Balasubramanyam, Joel B. Krier, Majid Mojarrad, Maria Francesca Bedeschi, Mahshid S. Azamian, Shruti Marwaha, Heather A. Colley, Katrina M. Dipple, Sirisak Chanprasert, Alexa T. McCray, Nicholas Stong, Anne V. Hing, Laura A. Mamounas, Edward C. Smith, Lauren C. Briere, John J.E. Mulvihill, Virginia P. Sybert, Maura R.Z. Ruzhnikov, Valerie Maduro, Frances A. High, Manish J. Butte, Willa Thorson, J. Carl Pallais, Jennefer N. Kohler, Dana Kiley, Raphael Bernier, Christina Lam, Michael J. Bamshad, Patricia A. Ward, Michael F. Wangler, Anita E. Beck, Shinya Yamamoto, Beverly Berg-Rood, Robb Rowley, Gabor T. Marth, Cynthia M. Cooper, Jeffrey G. Jarvik, Thomas C. Markello, Saskia Biskup, Devin Oglesbee, Laura Duncan, Elijah Kravets, Daniel J. Wegner, Mercedes E. Alejandro, Sarah K. Nicholas, Jennifer A. Wambach, Marni J. Falk, Brianna M. Tucker, Marie Morimoto, Corina Heller, Donna Novacic, Camilo Toro, Ashley Andrews, James P. Orengo, Shweta U. Dhar, and Pauline E. Schneeberger
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Death Domain Receptor Signaling Adaptor Proteins ,Programmed cell death ,Developmental Disabilities ,Biology ,03 medical and health sciences ,0302 clinical medicine ,Epidermal growth factor ,medicine ,Guanine Nucleotide Exchange Factors ,Humans ,Death domain ,Kinase ,Original Articles ,Phenotype ,Hypotonia ,Protein Transport ,030104 developmental biology ,Mutation ,Cancer research ,Human medicine ,Neurology (clinical) ,Nervous System Diseases ,Signal transduction ,medicine.symptom ,030217 neurology & neurosurgery ,Signal Transduction - Abstract
In pleiotropic diseases, multiple organ systems are affected causing a variety of clinical manifestations. Here, we report a pleiotropic disorder with a unique constellation of neurological, endocrine, exocrine, and haematological findings that is caused by biallelic MADD variants. MADD, the mitogen-activated protein kinase (MAPK) activating death domain protein, regulates various cellular functions, such as vesicle trafficking, activity of the Rab3 and Rab27 small GTPases, tumour necrosis factor-α (TNF-α)-induced signalling and prevention of cell death. Through national collaboration and GeneMatcher, we collected 23 patients with 21 different pathogenic MADD variants identified by next-generation sequencing. We clinically evaluated the series of patients and categorized the phenotypes in two groups. Group 1 consists of 14 patients with severe developmental delay, endo- and exocrine dysfunction, impairment of the sensory and autonomic nervous system, and haematological anomalies. The clinical course during the first years of life can be potentially fatal. The nine patients in Group 2 have a predominant neurological phenotype comprising mild-to-severe developmental delay, hypotonia, speech impairment, and seizures. Analysis of mRNA revealed multiple aberrant MADD transcripts in two patient-derived fibroblast cell lines. Relative quantification of MADD mRNA and protein in fibroblasts of five affected individuals showed a drastic reduction or loss of MADD. We conducted functional tests to determine the impact of the variants on different pathways. Treatment of patient-derived fibroblasts with TNF-α resulted in reduced phosphorylation of the extracellular signal-regulated kinases 1 and 2, enhanced activation of the pro-apoptotic enzymes caspase-3 and -7 and increased apoptosis compared to control cells. We analysed internalization of epidermal growth factor in patient cells and identified a defect in endocytosis of epidermal growth factor. We conclude that MADD deficiency underlies multiple cellular defects that can be attributed to alterations of TNF-α-dependent signalling pathways and defects in vesicular trafficking. Our data highlight the multifaceted role of MADD as a signalling molecule in different organs and reveal its physiological role in regulating the function of the sensory and autonomic nervous system and endo- and exocrine glands.
- Published
- 2020
14. Hypoglycemia in CDG patients due to PMM2 mutations: Follow up on hyperinsulinemic patients
- Author
-
Pascale de Lonlay, Jaak Jaeken, Carolyn Ellaway, Hossein Moravej, Saadet Mercimek-Andrews, Gregory M. Enns, David Coman, Shanti Balasubramaniam, Ruqaiah Altassan, Eva Morava, and Peter Witters
- Subjects
Research Report ,Pediatrics ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,lcsh:QH426-470 ,congenital disorder(s) of glycosylation ,Endocrinology, Diabetes and Metabolism ,PMM2‐CDG ,hyperinsulinism ,Hypoglycemia ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,Internal Medicine ,medicine ,Diazoxide ,In patient ,Prospective cohort study ,phosphomannomutase 2 ,lcsh:RC648-665 ,business.industry ,nutritional and metabolic diseases ,Research Reports ,medicine.disease ,diazoxide ,lcsh:Genetics ,hypoglycemia ,Etiology ,CDG ,Good prognosis ,business ,Hyperinsulinism ,Congenital disorder of glycosylation ,medicine.drug - Abstract
BACKGROUND: Phosphomannomutase 2 deficiency (PMM2-CDG) is the most common congenital disorder of glycosylation (CDG). Hypoglycemia has been reported in various CDG including PMM2-CDG. The frequency and etiology of hypoglycemia in PMM2-CDG are not well studied. METHODS: We conducted a systematic review of the literature on genetically and/or biochemically confirmed PMM2-CDG patients who developed hypoglycemia. Prospective follow-up information on the patients who received diazoxide therapy was collected and evaluated. RESULTS: A total of 165 peer-reviewed articles reporting on 933 PMM2-CDG patients were assessed. Hypoglycemia was specifically mentioned only in 23 of these patients (2.5%). Hyperinsulinism was identified in 10 patients (43% of all hypoglycemic patients). Among these 10 patients, seven were successfully treated with diazoxide. However, most patients remained on therapy longer than a year to stay free of hypoglycemia. CONCLUSION: Hypoglycemia is a rarely reported finding in patients with PMM2-CDG. Diazoxide-responsive hyperinsulinism was found to have a good prognosis on medication in our PMM2-CDG patients with hypoglycemia. No genotype-phenotype correlation was observed with respect to hyperinsulinism. A prospective study should be undertaken to explore the hypothesis that hypoglycemia is underdiagnosed in PMM2-CDG and to evaluate whether hyperinsulinism is always associated with hypoglycemia. ispartof: JIMD Rep vol:51 issue:1 pages:76-81 ispartof: location:United States status: Published online
- Published
- 2020
15. PMM2-CDG and nephrotic syndrome: A case report
- Author
-
Giuseppe Banderali, Elisabetta Salvatici, Valentina Rovelli, and Jaak Jaeken
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,nephrotic syndrome ,CDG ,PMM2‐CDG ,General Medicine ,renal involvement ,phosphomannomutase 2 deficiency ,congenital disorders of glycosylation - Abstract
Congenital disorders of glycosylation (CDG) are a group of rare metabolic diseases, characterized by a defect in the protein glycosylation process. Enzymes involved in this metabolic mechanism have ubiquitous distribution; thus, their alteration can cause systemic involvement and considerable phenotypic variability. Nephrotic syndrome (NS) is a clinical condition characterized by edema, hypoalbuminemia, hyperlipidemia, and proteinuria. We hereby report the case of a girl with central hypotonia, epilepsy, and severe psychomotor delay diagnosed with phosphomannomutase 2 deficiency (PMM2-CDG) after presenting with nephrotic syndrome at age 4 years. ispartof: CLINICAL CASE REPORTS vol:10 issue:2 ispartof: location:England status: published
- Published
- 2022
16. Disorders of Glutamine, Serine and Asparagine Metabolism
- Author
-
Jaak Jaeken, Johannes Häberle, and Olivier Dulac
- Subjects
Serine ,Glutamine ,Biochemistry ,Asparagine metabolism ,Glutamine synthetase ,Epileptic encephalopathy ,Serine racemase ,Biology - Published
- 2022
17. Congenital Disorders of Glycosylation
- Author
-
Jaak Jaeken and Lambert van den Heuvel
- Published
- 2022
18. COG6-CDG: Novel variants and novel malformation
- Author
-
Lara Cirnigliaro, Paolo Bianchi, Luisa Sturiale, Domenico Garozzo, Giovanna Mangili, Liesbeth Keldermans, Renata Rizzo, Gert Matthijs, Agata Fiumara, Jaak Jaeken, and Rita Barone
- Subjects
Embryology ,Glycosylation ,combined N- and O-glycosylation defect ,Health, Toxicology and Mutagenesis ,Golgi Apparatus ,congenital ano-rectal malformations ,CONGENITAL DISORDERS ,Toxicology ,PATIENT ,O-GLYCOSYLATION ,Congenital Disorders of Glycosylation ,Humans ,SUBUNIT-6 ,TRAFFICKING ,congenital disorder of glycosylation (CDG) ,MUTATION ,Science & Technology ,Vaginal Fistula ,Infant, Newborn ,N-GLYCOSYLATION ,GENE ,DEFICIENCY ,Adaptor Proteins, Vesicular Transport ,COG6 ,Pediatrics, Perinatology and Child Health ,Female ,OLIGOMERIC GOLGI-COMPLEX ,corpus callosum dysgenesis ,Life Sciences & Biomedicine ,Developmental Biology - Abstract
BACKGROUND: Deficiency of Conserved Oligomeric Golgi (COG) subunits (COG1-8) is characterized by both N- and O-protein glycosylation defects associated with destabilization and mislocalization of Golgi glycosylation machinery components (COG-CDG). Patients with COG defects present with neurological and multisystem involvement and possible malformation occurrence. Eighteen patients with COG6-CDG (COG6 mutations) were reported to date. We describe a patient with COG6-CDG with novel variants and a novel clinical feature namely a congenital recto-vaginal fistula. METHODS: In-depth serum N- and O-glycosylation structural analyses were conducted by MALDI-TOF mass spectrometry. COG6 variants were identified by a gene panel and confirmed by Sanger sequencing. RESULTS: This female newborn presented with facial dysmorphism, distal arthrogryposis and recurrent stool discharges per vaginam. A double-contrast barium-enema X-ray study revealed a dehiscence (approximately 5 mm) at the anterior wall of the rectal ampoule communicating with the vagina consistent with a recto-vaginal fistula. She had developmental delay, corpus callosum dysgenesis, liver and gastrointestinal involvement, hyperthermia episodes and early demise. Serum N- and O-glycosylation analyses pointed to a profound Golgi disarrangement. We identified two novel variants in COG6: a deletion of 1 bp mutation c.823delA creating a shift in the reading frame and a premature stop codon and a 3 bp deletion (c.1141_1143delCTC) producing an in-frame deletion of 1 amino acid. CONCLUSION: The congenital recto-vaginal fistula is a rare type of anorectal malformation that, to our knowledge, has not been reported in patients with a COG6 defect nor in patients with other COG defects. This study broadens COG6-CDG genetic landscape and spectrum of malformations. ispartof: BIRTH DEFECTS RESEARCH vol:114 issue:5-6 pages:165-174 ispartof: location:United States status: published
- Published
- 2022
19. Assessing the effects of PMM2 variants on protein stability
- Author
-
Dulce Quelhas, Sérgio F. Sousa, Jaak Jaeken, Mónica Lopes-Marques, S.S. Teixeira Carla, João Carneiro, Carlos Ferreira, J.F. Rocha, Esmeralda Martins, and Luísa Azevedo
- Subjects
Protein Conformation ,Protein Stability ,Endocrinology, Diabetes and Metabolism ,In silico ,Mutation, Missense ,Protein level ,Phosphomannomutase 2 deficiency ,Computational biology ,Biology ,Molecular Dynamics Simulation ,Pathogenicity ,Biochemistry ,Biochemical phenotype ,Endocrinology ,Protein stability ,Phosphotransferases (Phosphomutases) ,Genetics ,Amino acid residue ,Protein Multimerization ,Molecular Biology ,Relevant information - Abstract
Phosphomannomutase 2 deficiency, PMM2-CDG, is the most frequent disorder of protein N-glycosylation. It is an autosomal recessive disease with a broad clinical and biochemical phenotype. Trying to predict the impact of novel variants is often a challenge due to the high number of variants and the difficulty to establish solid genotype-phenotype correlations. A potential useful strategy is to use computational chemistry calculations as a tool from which relevant information on the structural impact of novel variants may be deduced. Here we present our analyses based on four well-known PMM2 deleterious variants (p.(Leu32Arg), p.(Asp65Tyr), p.(Phe119Leu), p.(Arg141His)) and the polymorphic p.(Glu197Ala) for which we have predicted the effect on protein stability. Our work predicts the effect of different amino acid residues on the conformation and stability of PMM2. These computational simulations are, therefore, an extremely useful methodology which, in combination with routinely used in silico methods of pathogenicity prediction, may help to reveal the structural impact of novel variants at the protein level, potentially leading to a better understanding of target biological molecules.
- Published
- 2021
20. ALG12-CDG: novel glycophenotype insights endorse the molecular defect
- Author
-
Luisa Sturiale, Agata Fiumara, Domenico Garozzo, Rita Barone, Alessandra Terracciano, Angelo Palmigiano, Francesca Esposito, Chiara Barone, Jaak Jaeken, Antonio Novelli, Emanuele Agolini, Angela Messina, and Sebastiano Bianca
- Subjects
Male ,congenital, hereditary, and neonatal diseases and abnormalities ,Glycan ,Glycosylation ,IgG ,Immunoglobulins ,Oligosaccharides ,Mannose ,Endoplasmic Reticulum ,Mannosyltransferases ,Biochemistry ,Abnormal glycosylation ,03 medical and health sciences ,chemistry.chemical_compound ,Congenital Disorders of Glycosylation ,Polysaccharides ,Glycophenotype ,Exome Sequencing ,ALG12-CDG ,Humans ,Immunodeficiency ,IgG Deficiency ,Child ,Molecular Biology ,Glycoproteins ,030304 developmental biology ,chemistry.chemical_classification ,0303 health sciences ,biology ,030302 biochemistry & molecular biology ,Transferrin ,Infant ,Cell Biology ,Molecular biology ,Blood proteins ,Novel variants ,carbohydrates (lipids) ,chemistry ,Child, Preschool ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,biology.protein ,Female ,Antibody ,Glycoprotein - Abstract
Congenital disorders of glycosylation (CDG) are genetic diseases characterized by deficient synthesis (CDG type I) and/or abnormal processing (CDG type II) of glycan moieties linked to protein and lipids. The impact of the molecular defects on protein glycosylation and in turn on the clinical phenotypes of patients with CDG is not yet understood. ALG12-CDG is due to deficiency of ALG12 α1,6-mannosyltransferase that adds the eighth mannose residue on the dolichol-PP-oligosaccharide precursor in the endoplasmic reticulum. ALG12-CDG is a severe multisystem disease associated with low to deficient serum immunoglobulins and recurrent infections. We thoroughly investigated the glycophenotype in a patient with novel ALG12 variants and immunodeficiency. We analyzed serum native transferrin, as first line test for CDG and we profiled serum IgG and total serum N-glycans by a combination of consolidated (N-glycan analysis by MALDI MS) and innovative mass spectrometry-based protocols, such as GlycoWorks RapiFluor N-glycan analysis coupled with LC-ESI MS. Intact serum transferrin showed, as expected for a CDG type I defect, underoccupancy of N-glycosylation sites. Surprisingly, total serum proteins and IgG N-glycans showed some specific changes, consisting in accumulating amounts of definite high-mannose and hybrid structures. As a whole, ALG12-CDG behaves as a dual CDG (CDG-I and II defects) and it is associated with distinct, abnormal glycosylation of total serum and IgG N-glycans. Glycan profiling of target glycoproteins may endorse the molecular defect unraveling the complex clinical phenotype of CDG patients.
- Published
- 2019
21. Long-term follow-up in PMM2-CDG: are we ready to start treatment trials?
- Author
-
Arnaud Bruneel, Eric Bauchart, Peter Witters, Wouter Meersseman, Gert Matthijs, Delphine Borgel, David Cassiman, Eva Morava, Tiffany Pascreau, Lonlay Pascale de, Tomas Honzik, Sandrine Vuillaumier, Jaak Jaeken, Nathalie Seta, and Ruqaiah Altassan
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Pediatrics ,medicine.medical_specialty ,medicine.diagnostic_test ,business.industry ,Antithrombin ,030105 genetics & heredity ,medicine.disease ,Clinical trial ,03 medical and health sciences ,030104 developmental biology ,Thyroid-stimulating hormone ,medicine ,Thyroid function ,Liver function tests ,Strabismus ,business ,Congenital disorder of glycosylation ,Genetics (clinical) ,Partial thromboplastin time ,medicine.drug - Abstract
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. 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. 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. 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.
- Published
- 2019
22. Mutations in MAGT1 lead to a glycosylation disorder with a variable phenotype
- Author
-
Eline Blommaert, Jaak Jaeken, Natalia A. Cherepanova, François Foulquier, Daisy Rymen, Reid Gilmore, Rebecca Sparkes, Romain Péanne, Anniek Corveleyn, Gert Matthijs, Valerie Race, Kaustuv Bhattacharya, Rik Schrijvers, Erika Souche, Christine Devalck, Frederik Staels, Liesbeth Keldermans, Molecular Diagnostics, Center for Human Genetics, Gasthuisberg, Katholieke Universiteit Leuven and Flanders Interuniversity Institute for Biotechnology 4, Leuven, Belgium, Catholic University of Leuven - Katholieke Universiteit Leuven (KU Leuven), Center for Human Genetics, Laboratory of clinical immunology, Unité de Glycobiologie Structurale et Fonctionnelle UMR 8576 (UGSF), Université de Lille-Institut National de la Recherche Agronomique (INRA)-Centre National de la Recherche Scientifique (CNRS), Université de Lille, CNRS, Unité de Glycobiologie Structurale et Fonctionnelle UMR 8576 [UGSF], Université de Lille-Centre National de la Recherche Scientifique (CNRS), Unité de Glycobiologie Structurale et Fonctionnelle - UMR 8576 (UGSF), and Université de Lille-Centre National de la Recherche Scientifique (CNRS)-Institut National de la Recherche Agronomique (INRA)
- Subjects
Male ,Glycosylation ,Adolescent ,[SDV]Life Sciences [q-bio] ,Protein subunit ,DNA Mutational Analysis ,congenital disorders of glycosylation ,CDG ,XMEN ,oligosaccharyltransferase complex ,Biology ,MAGT1 Deficiency ,03 medical and health sciences ,chemistry.chemical_compound ,Congenital Disorders of Glycosylation ,0302 clinical medicine ,medicine ,Humans ,Congenital disorders of glycosylation ,[SDV.BBM]Life Sciences [q-bio]/Biochemistry, Molecular Biology ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biochemistry [q-bio.BM] ,Child ,Cation Transport Proteins ,ComputingMilieux_MISCELLANEOUS ,030304 developmental biology ,Genetics ,0303 health sciences ,Multidisciplinary ,Tumor Suppressor Proteins ,Oligosaccharyltransferase ,Membrane Proteins ,Généralités ,Biological Sciences ,medicine.disease ,Phenotype ,[SDV.BBM.BC]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Biomolecules [q-bio.BM] ,3. Good health ,carbohydrates (lipids) ,Hexosyltransferases ,Oligosaccharyltransferase complex ,chemistry ,030220 oncology & carcinogenesis ,Primary immunodeficiency ,Lipid glycosylation - Abstract
Congenital disorders of glycosylation (CDG) are a group of rare metabolic diseases, due to impaired protein and lipid glycosylation. We identified two patients with defective serum transferrin glycosylation and mutations in the MAGT1 gene. These patients present with a phenotype that is mainly characterized by intellectual and developmental disability. MAGT1 has been described to be a subunit of the oligosaccharyltransferase (OST) complex and more specifically of the STT3B complex. However, it was also claimed that MAGT1 is a magnesium (Mg 2+ ) transporter. So far, patients with mutations in MAGT1 were linked to a primary immunodeficiency, characterized by chronic EBV infections attributed to a Mg 2+ homeostasis defect (XMEN). We compared the clinical and cellular phenotype of our two patients to that of an XMEN patient that we recently identified. All three patients have an N-glycosylation defect, as was shown by the study of different substrates, such as GLUT1 and SHBG, demonstrating that the posttranslational glycosylation carried out by the STT3B complex is dysfunctional in all three patients. Moreover, MAGT1 deficiency is associated with an enhanced expression of TUSC3, the homolog protein of MAGT1, pointing toward a compensatory mechanism. Hence, we delineate MAGT1-CDG as a disorder associated with two different clinical phenotypes caused by defects in glycosylation., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2019
23. AZATAX: Acetazolamide safety and efficacy in cerebellar syndrome in PMM2 congenital disorder of glycosylation (PMM2‐CDG)
- Author
-
E. Aísa, V. Freniche, Oscar Jerez García, Belén Pérez-Dueñas, N. Conde-Lorenzo, Celia Pérez-Cerdá, M.C. Miranda, Mercè Bolasell, Jordi Muchart, Manuel B. Morales, D. Itzep, F. Carratalá, I. Sebastián-García, Maria Eugenia Yoldi, E. de la Morena, S. Gassiot, L. López, María L. Couce, Jaak Jaeken, Sergio Aguilera-Albesa, Rafael Artuch, Raquel Montero, P. Arango, Luis González Gutiérrez-Solana, Dídac Casas-Alba, Daniel Cuadras, Mercedes Serrano, Susana Roldán, M.L. Carrasco-Marina, Antonio Martinez-Monseny, Alfons Macaya, Belén Pérez, Eduardo López-Laso, Javier Corral, L. Callejón-Póo, Ramón Velázquez-Fragua, M.C. Sierra-Córcoles, and Ramón Cancho-Candela
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Glycosylation ,Adolescent ,law.invention ,Young Adult ,03 medical and health sciences ,Epilepsy ,Congenital Disorders of Glycosylation ,0302 clinical medicine ,Randomized controlled trial ,Cerebellar Diseases ,law ,Internal medicine ,medicine ,Humans ,Single-Blind Method ,Carbonic Anhydrase Inhibitors ,Child ,Adverse effect ,Prothrombin time ,medicine.diagnostic_test ,business.industry ,medicine.disease ,Acetazolamide ,Treatment Outcome ,030104 developmental biology ,Neurology ,Phosphotransferases (Phosphomutases) ,Child, Preschool ,Female ,International Cooperative Ataxia Rating Scale ,Neurology (clinical) ,Lipodystrophy ,business ,Congenital disorder of glycosylation ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Objective: Phosphomannomutase deficiency (PMM2 congenital disorder of glycosylation [PMM2-CDG]) causes cerebellar syndrome and strokelike episodes (SLEs). SLEs are also described in patients with gain-of-function mutations in the CaV2.1 channel, for which acetazolamide therapy is suggested. Impairment in N-glycosylation of CaV2.1 promotes gain-of-function effects and may participate in cerebellar syndrome in PMM2-CDG. AZATAX was designed to establish whether acetazolamide is safe and improves cerebellar syndrome in PMM2-CDG. Methods: A clinical trial included PMM2-CDG patients, with a 6-month first-phase single acetazolamide therapy group, followed by a randomized 5-week withdrawal phase. Safety was assessed. The primary outcome measure was improvement in the International Cooperative Ataxia Rating Scale (ICARS). Other measures were the Nijmegen Pediatric CDG Rating Scale (NPCRS), a syllable repetition test (PATA test), and cognitive scores. Results: Twenty-four patients (mean age = 12.3 ± 4.5 years) were included, showing no serious adverse events. Thirteen patients required dose adjustment due to low bicarbonate or asthenia. There were improvements on ICARS (34.9 ± 23.2 vs 40.7 ± 24.8, effect size = 1.48, 95% confidence interval [CI] = 4.0–7.6, p < 0.001), detected at 6 weeks in 18 patients among the 20 responders, on NPCRS (95% CI = 0.3–1.6, p = 0.013) and on the PATA test (95% CI = 0.5–3.0, p = 0.006). Acetazolamide improved prothrombin time, factor X, and antithrombin. Clinical severity, epilepsy, and lipodystrophy predicted greater response. The randomized withdrawal phase showed ICARS worsening in the withdrawal group (effect size = 1.46, 95% CI = 2.65–7.52, p = 0.001). Interpretation: AZATAX is the first clinical trial of PMM2-CDG. Acetazolamide is well tolerated and effective for motor cerebellar syndrome. Its ability to prevent SLEs and its long-term effects on kidney function should be addressed in future studies. Ann Neurol 2019;85:740–751.
- Published
- 2019
24. A novel missense variant in SLC18A2 causes recessive brain monoamine vesicular transport disease and absent serotonin in platelets
- Author
-
Nihr BioResource, Jaak Jaeken, Lieven Lagae, Ernest Turro, Kate Downes, Kathleen Stirrups, Daniel Greene, Manisha Padmakumar, Chantal Thys, Kathleen Freson, Vincent Ramaekers, and Chris Van Geet
- Subjects
medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Case Report ,Case Reports ,QH426-470 ,Vesicular monoamine transporter 2 ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Diseases of the endocrine glands. Clinical endocrinology ,vesicular monoamine transporter 2 ,Neurodevelopmental disorder ,Internal medicine ,Internal Medicine ,medicine ,Genetics ,Missense mutation ,Platelet ,whole genome sequencing ,biology ,platelet dense granules ,business.industry ,medicine.disease ,RC648-665 ,Hypotonia ,serotonin ,Vesicular transport protein ,Monoamine neurotransmitter ,Endocrinology ,biology.protein ,epilepsy ,Serotonin ,medicine.symptom ,business - Abstract
BACKGROUND: Brain monoamine vesicular transport disease is an infantile onset neurodevelopmental disorder caused by variants in SLC18A2, which codes for the vesicular monoamine transporter 2 (VMAT2) protein, involved in the transport of monoamines into synaptic vesicles and of serotonin into platelet dense granules. CASE PRESENTATION: The presented case is of a child, born of healthy consanguineous parents, who exhibited hypotonia, mental disability, epilepsy, uncontrolled movements, and gastrointestinal problems. A trial treatment with L-DOPA proved unsuccessful and the exact neurological involvement could not be discerned due to normal metabolic and brain magnetic resonance imaging results.Platelet studies and whole genome sequencing were performed. At age 4, the child's platelets showed a mild aggregation and adenosine triphosphate secretion defect that could be explained by dysmorphic dense granules observed by electron microscopy. Interestingly, the dense granules were almost completely depleted of serotonin. A novel homozygous p.P316A missense variant in VMAT2 was detected in the patient and the consanguineous parents were found to be heterozygous for this variant. Although the presence of VMAT2 on platelet dense granules has been demonstrated before, this is the first report of defective platelet dense granule function related to absent serotonin storage in a patient with VMAT2 deficiency but without obvious clinical bleeding problems. CONCLUSIONS: This study illustrates the homology between serotonin metabolism in brain and platelets, suggesting that these blood cells can be model cells for some pathways relevant for neurological diseases. The literature on VMAT2 deficiency is reviewed. ispartof: JIMD Rep vol:47 issue:1 pages:9-16 ispartof: location:United States status: Published online
- Published
- 2019
25. Clinical Utility Gene Card for: PGM3 defective congenital disorder of glycosylation
- Author
-
Jaak Jaeken, Dirk J. Lefeber, and Gert Matthijs
- Subjects
Congenital Disorders of Glycosylation ,Genotype ,Phosphoglucomutase ,Clinical Utility Gene Card ,Mutation ,Genetics ,Humans ,Genetic Predisposition to Disease ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Risk Assessment ,Sensitivity and Specificity ,Genetics (clinical) - Abstract
Contains fulltext : 215589.pdf (Publisher’s version ) (Closed access) Review of the analytical and clinical validity as well as of the clinical utility of DNA-based testing for mutations in PGM3 in diagnostic, predictive and prenatal settings, and for risk assessment in relatives.
- Published
- 2019
26. Keeping an eye on congenital disorders of O‐glycosylation: A systematic literature review
- Author
-
David Coman, Jaak Jaeken, Vanessa dos Reis Ferreira, Dorinda Marques-da-Silva, Carlota Pascoal, Eva Morava, Glen A. Gole, and Rita Francisco
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Pediatrics ,medicine.medical_specialty ,Glycosylation ,Eye Diseases ,macromolecular substances ,03 medical and health sciences ,chemistry.chemical_compound ,Congenital Disorders of Glycosylation ,0302 clinical medicine ,Genetics ,medicine ,Animals ,Humans ,In patient ,Genetics (clinical) ,business.industry ,Human genetics ,carbohydrates (lipids) ,030104 developmental biology ,Systematic review ,chemistry ,Eye disorder ,lipids (amino acids, peptides, and proteins) ,business ,030217 neurology & neurosurgery - Abstract
Congenital disorders of glycosylation (CDG) are a rapidly growing family comprising >100 genetic diseases. Some 25 CDG are pure O-glycosylation defects. Even among this CDG subgroup, phenotypic diversity is broad, ranging from mild to severe poly-organ/system dysfunction. Ophthalmic manifestations are present in 60% of these CDG. The ophthalmic manifestations in N-glycosylation-deficient patients have been described elsewhere. The present review documents the spectrum and incidence of eye disorders in patients with pure O-glycosylation defects with the aim of assisting diagnosis and management and promoting research.
- Published
- 2019
27. A Patient with neonatal cholestasis
- Author
-
Kristl G, Claeys, Luc, Breysem, Eric, Legius, Hilde, Brems, David, Cassiman, Matthieu, Moisse, Pieter, Vermeersch, Elena, Levtchenko, and Jaak, Jaeken
- Subjects
musculoskeletal diseases ,Adult ,Male ,Gilbert syndrome ,Cholestasis ,Case Study ,Infant, Newborn ,Fibrous Dysplasia, Polyostotic ,polyostotic fibrous dysplasia ,Infant, Newborn, Diseases ,CAT-2 ,Charcot-Marie-Tooth Disease ,Mutation ,Humans ,dynamin-2 deficiency - Abstract
The patient, a boy born in 1991, showed pronounced polyostotic fibrous dysplasia due to McCune–Albright syndrome, as well as Gilbert syndrome and Charcot–Marie–Tooth neuropathy caused by a DNM2 mutation. In addition, the patient, his sister, mother and maternal grandfather had intermittently increased plasma arginine and lysine levels, most probably due to heterozygosity for a novel pathogenic SLC7A2 variant.
- Published
- 2021
28. Aberrant sialylation in a patient with a HNF1α variant and liver adenomatosis
- Author
-
Rita Barone, Angelo Palmigiano, Domenico Garozzo, Nicole Revencu, Jaak Jaeken, Immacolata Speciale, Rosangela Artuso, Gert Matthijs, Xavier Stéphenne, Gaetano Bertino, Marie-Cécile Nassogne, Luisa Sturiale, Cristina De Castro, Angela Messina, Sturiale, Luisa, Nassogne, Marie-Cécile, Palmigiano, Angelo, Messina, Angela, Speciale, Immacolata, Artuso, Rosangela, Bertino, Gaetano, Revencu, Nicole, Stephénne, Xavier, De Castro, Cristina, Matthijs, Gert, Barone, Rita, Jaeken, Jaak, Garozzo, Domenico, UCL - SSS/IREC/PEDI - Pôle de Pédiatrie, UCL - (SLuc) Service de neurologie pédiatrique, UCL - (SLuc) Service de gastro-entérologie et hépatologie pédiatrique, UCL - SSS/IREC/SLUC - Pôle St.-Luc, UCL - SSS/IONS/NEUR - Clinical Neuroscience, and UCL - (SLuc) Centre de génétique médicale UCL
- Subjects
0301 basic medicine ,Proteomics ,Glycosylation ,Science ,02 engineering and technology ,Biology ,Article ,Glycomics ,03 medical and health sciences ,chemistry.chemical_compound ,Glycosylation analysis ,Biological Sciences ,chemistry.chemical_classification ,Multidisciplinary ,Hypersialylated N-glycans ,Isoelectric focusing ,Biological Science ,021001 nanoscience & nanotechnology ,Phenotype ,Sialic acid ,carbohydrates (lipids) ,030104 developmental biology ,chemistry ,Biochemistry ,Transferrin ,Glycomic ,0210 nano-technology ,Glycoprotein ,HNF1 alpha mutated HCA - Abstract
Summary Glycosylation is a fundamental post-translational modification of proteins that boosts their structural diversity providing subtle and specialized biological properties and functions. All those genetic diseases due to a defective glycan biosynthesis and attachment to the nascent glycoproteins fall within the wide area of congenital disorders of glycosylation (CDG), mostly causing multisystem involvement. In the present paper, we detailed the unique serum N-glycosylation of a CDG-candidate patient with an unexplained neurological phenotype and liver adenomatosis harboring a recurrent pathogenic HNF1α variant. Serum transferrin isoelectric focusing showed a surprising N-glycosylation pattern consisting on hyposialylation, as well as remarkable hypersialylation. Mass spectrometry-based glycomic analyses of individual serum glycoproteins enabled to unveil hypersialylated complex N-glycans comprising up to two sialic acids per antenna. Further advanced MS analysis showed the additional sialic acid is bonded through an α2-6 linkage to the peripheral N-acetylglucosamine residue., Graphical abstract, Highlights • Serum N-glycome is altered in a boy with neurological syndrome and HNF1α mutated HCA • Glycomics reveals unique hypersialylated N-glycans with two NeuAc per antenna • In-depth MS studies show the additional NeuAc is α2-6 linked to an outer arm GlcNAc, Biological Sciences ; Proteomics ; Glycomics
- Published
- 2021
29. SLC37A4‐CDG : Second patient
- Author
-
Domenico Garozzo, François Foulquier, Matthew P. Wilson, Jaak Jaeken, Teresa Campos, Gert Matthijs, Valerie Race, Emile Van Schaftingen, Daisy Rymen, Elisa Leão-Teles, Liesbeth Keldermans, Erika Souche, Luisa Sturiale, Dulce Quelhas, Esmeralda Rodrigues, Unité de Glycobiologie Structurale et Fonctionnelle UMR 8576 (UGSF), Institut National de la Recherche Agronomique (INRA)-Université de Lille-Centre National de la Recherche Scientifique (CNRS), Université de Lille, CNRS, Unité de Glycobiologie Structurale et Fonctionnelle (UGSF) - UMR 8576, and Université de Lille-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Research Report ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Glycosylation ,lcsh:QH426-470 ,glycosylation ,Endocrinology, Diabetes and Metabolism ,[SDV]Life Sciences [q-bio] ,medicine.disease_cause ,Biochemistry, Genetics and Molecular Biology (miscellaneous) ,Gastroenterology ,lcsh:Diseases of the endocrine glands. Clinical endocrinology ,03 medical and health sciences ,Liver disease ,chemistry.chemical_compound ,CDG ,G6PT1 ,glycogen storage disease ,hepatopathy ,SLC37A4 ,Internal medicine ,Membranoproliferative glomerulonephritis ,Internal Medicine ,medicine ,Glycogen storage disease ,Mild dysmorphism ,030304 developmental biology ,0303 health sciences ,Type 1 diabetes ,Mutation ,lcsh:RC648-665 ,business.industry ,030305 genetics & heredity ,Research Reports ,medicine.disease ,3. Good health ,lcsh:Genetics ,chemistry ,business ,Congenital disorder of glycosylation - Abstract
Recently, a disorder caused by the heterozygous de novo c.1267C>T (p.R423*) substitution in SLC37A4 has been described. This causes mislocalization of the glucose-6-phosphate transporter to the Golgi leading to a congenital disorder of glycosylation type II (SLC37A4-CDG). Only one patient has been reported showing liver disease that improved with age and mild dysmorphism. Here we report the second patient with a type II CDG caused by the same heterozygous de novo c.1267C>T (p.R423*) mutation thereby confirming the pathogenicity of this variant and expanding the clinical picture with type 1 diabetes, severe scoliosis, and membranoproliferative glomerulonephritis. Additional clinical and biochemical data provide further insight into the mechanism and prognosis of SLC37A4-CDG. keywords: CDG, G6PT1, glycogen storage disease, glycosylation, hepatopathy, SLC37A4 ispartof: JIMD Reports vol:58 issue:1 pages:122-128 ispartof: location:United States status: published
- Published
- 2021
30. Clinical and radiological correlates of activities of daily living in cerebellar atrophy caused by PMM2 mutations (PMM2-CDG)
- Author
-
Renata Rizzo, Roberta Battini, Rossella Parini, Rita Barone, Gert Matthijs, Daniele Frattini, Domenico Garozzo, Elisa Biamino, Serena Gasperini, Giuseppe Sortino, Jaak Jaeken, Annalisa Madeo, Diego Martinelli, Maja Di Rocco, Fabio Pettinato, Agata Fiumara, Luisa Sturiale, F Sirchia, Giovanni Mostile, and Amelia Morrone
- Subjects
Male ,medicine.medical_specialty ,Cerebellum ,Ataxia ,Neurology ,Pons atrophy ,Activities of daily living ,Cerebellar atrophy ,Congenital disorder(s) of glycosylation ,PMM2 variants ,03 medical and health sciences ,Congenital Disorders of Glycosylation ,0302 clinical medicine ,Cerebellar Diseases ,Internal medicine ,Intellectual disability ,medicine ,Humans ,0303 health sciences ,business.industry ,030305 genetics & heredity ,medicine.disease ,Peripheral neuropathy ,medicine.anatomical_structure ,Phosphotransferases (Phosphomutases) ,Mutation ,Cerebellar vermis ,Original Article ,Neurology (clinical) ,Atrophy ,medicine.symptom ,business ,Congenital disorder of glycosylation ,030217 neurology & neurosurgery - Abstract
We aimed to identify clinical, molecular and radiological correlates of activities of daily living (ADL) in patients with cerebellar atrophy caused by PMM2 mutations (PMM2-CDG), the most frequent congenital disorder of glycosylation. Twenty-six PMM2-CDG patients (12 males; mean age 13 ± 11.1 years) underwent a standardized assessment to measure ADL, ataxia (brief ataxia rating scale, BARS) and phenotype severity (Nijmegen CDG rating scale, NCRS). MRI biometry of the cerebellum and the brainstem were performed in 23 patients (11 males; aged 5 months–18 years) and 19 control subjects with equal gender and age distributions. The average total ADL score was 15.3 ± 8.5 (range 3–32 out of 36 indicating severe functional disability), representing variable functional outcome in PMM2-CDG patients. Total ADL scores were significantly correlated with NCRS (r2 = 0.55, p < 0.001) and BARS scores (r2 = 0.764; p < 0.001). Severe intellectual disability, peripheral neuropathy, and severe PMM2 variants were all significantly associated with worse functional outcome. Higher ADL scores were significantly associated with decreased diameters of cerebellar vermis (r2 = 0.347; p = 0.004), hemispheres (r2 = 0.436; p = 0.005), and brainstem, particularly the mid-pons (r2 = 0.64; p < 0.001) representing the major radiological predictor of functional disability score in multivariate regression analysis. We show that cerebellar syndrome severity, cognitive level, peripheral neuropathy, and genotype correlate with ADL used to quantify disease-related deficits in PMM2-CDG. Brainstem involvement should be regarded among functional outcome predictors in patients with cerebellar atrophy caused by PMM2-CDG.
- Published
- 2021
31. D-galactose supplementation in individuals with PMM2-CDG: results of a multicenter, open label, prospective pilot clinical trial
- Author
-
Jaak Jaeken, David Cassiman, Clara D.M. van Karnebeek, Dirk Lefeber, Peter Witters, Eva Morava, Laura A. Tseng, and Hans C. Andersson
- Subjects
medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Glycosylation ,Pharmacology toxicology ,D-galactose ,lcsh:Medicine ,Nijmegen pediatric CDG rating scale (NPCRS) ,PMM2-CDG ,chemistry.chemical_compound ,Congenital Disorders of Glycosylation ,Internal medicine ,medicine ,Humans ,Pharmacology (medical) ,Prospective Studies ,Pmm2 cdg ,Trial registration ,Letter to the Editor ,Genetics (clinical) ,Congenital disorder of glycosylation (CDG) ,business.industry ,lcsh:R ,Pilot trial ,Galactose ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,General Medicine ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,medicine.disease ,Clinical trial ,chemistry ,Phosphotransferases (Phosphomutases) ,Dietary Supplements ,Open label ,business ,Congenital disorder of glycosylation - Abstract
PMM2-CDG is the most prevalent congenital disorder of glycosylation (CDG) with only symptomatic therapy. Some CDG have been successfully treated with D-galactose. We performed an open-label pilot trial with D-galactose in 9 PMM2-CDG patients. Overall, there was no significant improvement but some milder patients did show positive clinical changes; also there was a trend toward improved glycosylation. Larger placebo-controlled studies are required to determine whether D-galactose could be used as supportive treatment in PMM2-CDG patients.Trial registration ClinicalTrials.gov Identifier: NCT02955264. Registered 4 November 2016, https://clinicaltrials.gov/ct2/show/NCT02955264
- Published
- 2021
32. An international classification of inherited metabolic disorders (ICIMD)
- Author
-
Ferreira C. R., Rahman S., Keller M., Zschocke J., ICIMD Advisory Group: Jose Abdenur, Houda Ali, Rafael Artuch, Andrea Ballabio, Bruce Barshop, Matthias Baumgartner, Enrico Silvio Bertini, Nenad Blau, Valerio Carelli, Christopher Carroll, Patrick F Chinnery, John Christodoulou, Veronica Cornejo, Niklas Darin, Terry Derks, Daria Diodato, Carlo Dionisi-Vici, John A Duley, Toshi Fukao, Ángeles García-Cazorla, Roberto Giugliani, Amy Goldstein, Georg Hoffmann, Rita Horvath, Isabel Ibarra, Anita Inwood, Jaak Jaeken, Cecilia Jimenez-Mallebrera, Amel Karaa, Thomas Klopstock, Stefan Kölker, Cornelia Kornblum, Viktor Kožich, Costanza Lamperti, Nils-Göran Larsson, Aida Lemes, Barry Lewis, Michelangelo Mancuso, Robert McFarland, Fanny Mochel, Julio Montoya, Eva Morava, Karin Naess, Torayuki Okuyama, Annie Olry, Veronique Paquis-Flucklinger, Sumit Parikh, Marc Patterson, Ceila Pérez de Ferrán, Verena Peters, Holger Prokisch, Ann Saada, Gajja S Salomons, Jean-Marie Saudubray, Maurizio Scarpa, Ulrike Schara-Schmidt, Manuel Schiff, Serenella Servidei, Jan Smeitink, Anu Suomalainen, Trine Tangeraas, Robert W Taylor, Ines Thiele, David Thorburn, Johan Van Hove, Ans T Van der Ploeg, Clara Van Karnebeek, Gepke Visser, Jerry Vockley, Ronald Wanders, Dianne Webster, Anna Wedell, Veronica Wiley, Anna Wredenberg, Massimo Zeviani, C. R., Ferreira, S., Rahman, M., Keller, J., Zschocke, Advisory Group: Jose Abdenur, Icimd, Ali, Houda, Artuch, Rafael, Ballabio, Andrea, Barshop, Bruce, Baumgartner, Matthia, Silvio Bertini, Enrico, Blau, Nenad, Carelli, Valerio, Carroll, Christopher, F Chinnery, Patrick, Christodoulou, John, Cornejo, Veronica, Darin, Nikla, Derks, Terry, Diodato, Daria, Dionisi-Vici, Carlo, A Duley, John, Fukao, Toshi, García-Cazorla, Ángele, Giugliani, Roberto, Goldstein, Amy, Hoffmann, Georg, Horvath, Rita, Ibarra, Isabel, Inwood, Anita, Jaeken, Jaak, Jimenez-Mallebrera, Cecilia, Karaa, Amel, Klopstock, Thoma, Kölker, Stefan, Kornblum, Cornelia, Kožich, Viktor, Lamperti, Costanza, Larsson, Nils-Göran, Lemes, Aida, Lewis, Barry, Mancuso, Michelangelo, Mcfarland, Robert, Mochel, Fanny, Montoya, Julio, Morava, Eva, Naess, Karin, Okuyama, Torayuki, Olry, Annie, Paquis-Flucklinger, Veronique, Parikh, Sumit, Patterson, Marc, Pérez de Ferrán, Ceila, Peters, Verena, Prokisch, Holger, Saada, Ann, S Salomons, Gajja, Saudubray, Jean-Marie, Scarpa, Maurizio, Schara-Schmidt, Ulrike, Schiff, Manuel, Servidei, Serenella, Smeitink, Jan, Suomalainen, Anu, Tangeraas, Trine, W Taylor, Robert, Thiele, Ine, Thorburn, David, Van Hove, Johan, T Van der Ploeg, An, Van Karnebeek, Clara, Visser, Gepke, Vockley, Jerry, Wanders, Ronald, Webster, Dianne, Wedell, Anna, Wiley, Veronica, Wredenberg, Anna, Zeviani, Massimo, University of Zurich, Ferreira C.R., Rahman S., Keller M., Zschocke J., ICIMD Advisory Group, and Carelli V.
- Subjects
Nosology ,medicine.medical_specialty ,2716 Genetics (clinical) ,Expert advice ,610 Medicine & health ,Genetic Condition ,Article ,ICIMD ,03 medical and health sciences ,1311 Genetics ,International Classification of Diseases ,Genetics ,Humans ,Relevance (law) ,Medicine ,ontology ,Intensive care medicine ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,Mechanism (biology) ,business.industry ,030305 genetics & heredity ,inherited metabolic disorder ,classification ,inherited metabolic disorders ,10036 Medical Clinic ,Metabolism, Inborn Error ,business ,Metabolism, Inborn Errors ,Human - Abstract
Several initiatives at establishing a classification of inherited metabolic disorders have been published previously, some focusing on pathomechanisms, others on clinical manifestations, while yet another attempted a simplified approach of a comprehensive nosology. Some of these classifications suffered from shortcomings, such as lack of a mechanism for continuous update in light of a rapidly evolving field, or lack of widespread input from the metabolic community at large. Our classification-the International Classification of Inherited Metabolic Disorders, or International Classification of Inborn Metabolic Disorders (ICIMD)-includes 1450 disorders, and differs from prior approaches in that it benefited from input by a large number of experts in the field, and was endorsed by major metabolic societies around the globe. Several criteria such as pathway involvement and pathomechanisms were considered. The main purpose of the hierarchical, group-based approach of the ICIMD is an improved understanding of the interconnections between many individual conditions that may share functional, clinical, and diagnostic features. The ICIMD aims to include any primary genetic condition in which alteration of a biochemical pathway is intrinsic to specific biochemical, clinical, and/or pathophysiological features. As new disorders are discovered, we will seek the opinion of experts in the advisory board prior to inclusion in the appropriate group of the ICIMD, thus guaranteeing the continuing relevance of this classification via regular curation and expert advice.
- Published
- 2021
33. International consensus guidelines for phosphoglucomutase 1 deficiency (PGM1‐CDG ): Diagnosis, follow‐up, and management
- Author
-
Jaak Jaeken, Kristina Falkenstein, Carlos Ferreira, Andrew C. Edmondson, Paula A. Videira, Joy Lee, David Coman, Rita Barone, Tomas Honzik, Vanessa dos Reis Ferreira, Frederic Tort, Rita Francisco, Anna Cechova, Christina Lam, Mari Anne Vals, Hudson H. Freeze, Dorinda Marques-da-Silva, Nicol C. Voermans, Ruqaiah Altassan, Małgorzata Seroczyńska, Daisy Rymen, Dulce Quelhas, Carlota Pascoal, Sarah Donoghue, Peter Witters, Eva Morava, Donna M. Krasnewich, Jolanta Sykut-Cegielska, Sandra Brasil, Dirk Lefeber, Stephanie Grunewald, Christian Thiel, Mercedes Serrano, Agata Fiumara, Silvia Radenkovic, and Kimiyo Raymond
- Subjects
Adult ,Pediatrics ,medicine.medical_specialty ,congenital, hereditary, and neonatal diseases and abnormalities ,Consensus ,International Cooperation ,Cardiomyopathy ,Hypoglycemia ,Article ,d-galactose ,congenital disorder of glycosylation ,03 medical and health sciences ,Muscular Diseases ,phosphoglucomutase 1 deficiency ,PGM1 ,Health care ,Genetics ,medicine ,Glycogen storage disease ,Humans ,management guidelines ,Genetics (clinical) ,030304 developmental biology ,0303 health sciences ,business.industry ,030305 genetics & heredity ,PGM1-CDG ,Genetic disorder ,Disease Management ,Galactose ,Infant ,medicine.disease ,Glycogen Storage Disease ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Cleft Palate ,Liver function ,business ,Cardiomyopathies ,Congenital disorder of glycosylation - Abstract
Phosphoglucomutase 1 (PGM1) deficiency is a rare genetic disorder that affects glycogen metabolism, glycolysis, and protein glycosylation. Previously known as GSD XIV, it was recently reclassified as a congenital disorder of glycosylation, PGM1-CDG. PGM1-CDG usually manifests as a multisystem disease. Most patients present as infants with cleft palate, liver function abnormalities and hypoglycemia, but some patients present in adulthood with isolated muscle involvement. Some patients develop life-threatening cardiomyopathy. Unlike most other CDG, PGM1-CDG has an effective treatment option, d-galactose, which has been shown to improve many of the patients' symptoms. Therefore, early diagnosis and initiation of treatment for PGM1-CDG patients are crucial decisions. In this article, our group of international experts suggests diagnostic, follow-up, and management guidelines for PGM1-CDG. These guidelines are based on the best available evidence-based data and experts' opinions aiming to provide a practical resource for health care providers to facilitate successful diagnosis and optimal management of PGM1-CDG patients. ispartof: Journal Of Inherited Metabolic Disease vol:s44 issue:1 pages:148-163 ispartof: location:United States status: published
- Published
- 2021
34. Platelets and Defective N-Glycosylation
- Author
-
Elmina Mammadova-Bach, Attila Braun, Thomas Gudermann, and Jaak Jaeken
- Subjects
0301 basic medicine ,Blood Platelets ,Glycan ,congenital, hereditary, and neonatal diseases and abnormalities ,Glycosylation ,N-glycans ,Review ,030204 cardiovascular system & hematology ,Models, Biological ,Catalysis ,lcsh:Chemistry ,Inorganic Chemistry ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,N-linked glycosylation ,congenital disorders of N-glycosylation ,Animals ,Homeostasis ,Humans ,Platelet ,Asparagine ,Physical and Theoretical Chemistry ,lcsh:QH301-705.5 ,Molecular Biology ,Spectroscopy ,thrombosis ,biology ,Endoplasmic reticulum ,Organic Chemistry ,General Medicine ,Sequon ,Computer Science Applications ,Cell biology ,carbohydrates (lipids) ,030104 developmental biology ,lcsh:Biology (General) ,lcsh:QD1-999 ,chemistry ,platelets ,biology.protein ,hemostasis ,Protein folding ,lipids (amino acids, peptides, and proteins) ,Calcium ,Energy Metabolism ,megakaryopoiesis - Abstract
N-glycans are covalently linked to an asparagine residue in a simple acceptor sequence of proteins, called a sequon. This modification is important for protein folding, enhancing thermodynamic stability, and decreasing abnormal protein aggregation within the endoplasmic reticulum (ER), for the lifetime and for the subcellular localization of proteins besides other functions. Hypoglycosylation is the hallmark of a group of rare genetic diseases called congenital disorders of glycosylation (CDG). These diseases are due to defects in glycan synthesis, processing, and attachment to proteins and lipids, thereby modifying signaling functions and metabolic pathways. Defects in N-glycosylation and O-glycosylation constitute the largest CDG groups. Clotting and anticlotting factor defects as well as a tendency to thrombosis or bleeding have been described in CDG patients. However, N-glycosylation of platelet proteins has been poorly investigated in CDG. In this review, we highlight normal and deficient N-glycosylation of platelet-derived molecules and discuss the involvement of platelets in the congenital disorders of N-glycosylation. ispartof: INTERNATIONAL JOURNAL OF MOLECULAR SCIENCES vol:21 issue:16 ispartof: location:Switzerland status: published
- Published
- 2020
35. NGLY1 deficiency: Novel variants and literature review
- Author
-
Ariana Kariminejad, Jaak Jaeken, Hossein Najmabadi, Raoul C.M. Hennekam, Marjan Shakiba, Said Talebi, Maryam Eghbali, Mina Makvand, Mehrvash Shams, and Parva Namiranian
- Subjects
Adult ,Male ,Adolescent ,Hyperlordosis ,Hypotonia ,Bioinformatics ,Alacrimia ,Contractures ,Congenital Disorders of Glycosylation ,Protein Domains ,Intellectual disability ,Genetics ,medicine ,NGLY1 ,Humans ,Peptide-N4-(N-acetyl-beta-glucosaminyl) Asparagine Amidase ,Genetics (clinical) ,Muscle contracture ,Muscular hypotonia ,medicine.diagnostic_test ,business.industry ,General Medicine ,medicine.disease ,Muscle atrophy ,Pedigree ,Phenotype ,Congenital disorders of de-glycosylation ,Liver biopsy ,Protein deglycosylation ,Mutation ,Female ,medicine.symptom ,business - Abstract
NGLY1 deficiency is a recently described autosomal recessive disorder, involved in deglycosylation of proteins, and for that reason grouped as the congenital disorders of deglycosylation together with the lysosomal storage disorders. The typical phenotype is characterized by intellectual disability, liver malfunctioning, muscular hypotonia, involuntary movements, and decreased or absent tear production. Liver biopsy demonstrates vacuolar amorphous cytoplasmic storage material. NGLY1 deficiency is caused by bi-allelic variants in NGLY1 which catalyzes protein deglycosylation. We describe five patients from two families with NGLY1 deficiency due to homozygosity for two novel NGLY1 variants, and compare their findings to those of earlier reported patients. The typical features of the disorder are present in a limited way, and there is intra-familial variability. In addition in one of the families the muscle atrophy and posture abnormalities are marked. These can be explained either as variability of the phenotype or as sign of slowly progression of features as the present affected individuals are older than earlier reported patients.
- Published
- 2020
36. De novo loss-of-function variants in X-linked MED12 are associated with Hardikar syndrome in females
- Author
-
Dong, Li, Alanna, Strong, Kaitlyn M, Shen, David, Cassiman, Maria, Van Dyck, Natalia Duarte, Linhares, Eugenia Ribeiro, Valadares, Tiancheng, Wang, Sergio D J, Pena, Jaak, Jaeken, Samantha, Vergano, Elaine, Zackai, Anne, Hing, Penny, Chow, Arupa, Ganguly, Tasja, Scholz, Tatjana, Bierhals, Deindl, Philipp, Hakon, Hakonarson, and Elizabeth, Bhoj
- Subjects
Adult ,Cleft Palate ,Young Adult ,Cholestasis ,Mediator Complex ,Phenotype ,Genes, X-Linked ,Intellectual Disability ,Mental Retardation, X-Linked ,Humans ,Female ,Retinitis Pigmentosa - Abstract
Hardikar syndrome (MIM 612726) is a rare multiple congenital anomaly syndrome characterized by facial clefting, pigmentary retinopathy, biliary anomalies, and intestinal malrotation, but with preserved cognition. Only four patients have been reported previously, and none had a molecular diagnosis. Our objective was to identify the genetic basis of Hardikar syndrome (HS) and expand the phenotypic spectrum of this disorder.We performed exome sequencing on two previously reported and five unpublished female patients with a clinical diagnosis of HS. X-chromosome inactivation (XCI) studies were also performed.We report clinical features of HS with previously undescribed phenotypes, including a fatal unprovoked intracranial hemorrhage at age 21. We additionally report the discovery of de novo pathogenic nonsense and frameshift variants in MED12 in these seven individuals and evidence of extremely skewed XCI in all patients with informative testing.Pathogenic missense variants in the X-chromosome gene MED12 have previously been associated with Opitz-Kaveggia syndrome, Lujan syndrome, Ohdo syndrome, and nonsyndromic intellectual disability, primarily in males. We propose a fifth, female-specific phenotype for MED12, and suggest that nonsense and frameshift loss-of-function MED12 variants in females cause HS. This expands the MED12-associated phenotype in females beyond intellectual disability.
- Published
- 2020
37. New Insights into Immunological Involvement in Congenital Disorders of Glycosylation (CDG) from a People-Centric Approach
- Author
-
Jaak Jaeken, Paula A. Videira, Vanessa dos Reis Ferreira, Fernando Pimentel-Santos, Sandra Brasil, Rita Francisco, Carlota Pascoal, Dorinda Marques-da-Silva, Ana Rita Grosso, Ruqaiah Altassan, DCV - Departamento de Ciências da Vida, UCIBIO - Applied Molecular Biosciences Unit, Centro de Estudos de Doenças Crónicas (CEDOC), and NOVA Medical School|Faculdade de Ciências Médicas (NMS|FCM)
- Subjects
Pediatrics ,medicine.medical_specialty ,Allergy ,congenital, hereditary, and neonatal diseases and abnormalities ,Population ,QUESTIONNAIRE ,lcsh:Medicine ,BROAD-SPECTRUM ,macromolecular substances ,OSTEOPOROSIS ,e-questionnaire ,PATIENT ,Article ,PMM2-CDG ,immune response ,03 medical and health sciences ,Medicine, General & Internal ,0302 clinical medicine ,Immune system ,SDG 3 - Good Health and Well-being ,General & Internal Medicine ,VALPROIC ACID ,medicine ,infections ,Pmm2 cdg ,education ,people-centricity ,gastrointestinal tract (GI) ,030304 developmental biology ,0303 health sciences ,education.field_of_study ,Science & Technology ,IA ,business.industry ,allergies ,congenital disorder(s) of glycosylation (CDG) ,lcsh:R ,General Medicine ,CARE ,medicine.disease ,vaccination ,ANTICONVULSANT DRUG ,DEFICIENCY ,Vaccination ,gastrointestinal tract (GI), e-questionnaire ,business ,Life Sciences & Biomedicine ,030217 neurology & neurosurgery ,Red flags - Abstract
Congenital disorders of glycosylation (CDG) are rare diseases with variable phenotypes and severity. Immunological involvement remains a largely uncharted topic in CDG, mainly due to lack of robust data. To better characterize immune-related manifestations&rsquo, prevalence, relevance, and quality-of-life (QoL) impact, we developed electronic questionnaires targeting (1) CDG patients and (2) the general &ldquo, healthy&rdquo, population. Two-hundred and nine CDG patients/caregivers and 349 healthy participants were included in this study. PMM2-CDG was the most represented CDG (n = 122/209). About half of these participants (n = 65/122) described relevant infections with a noteworthy prevalence of those affecting the gastrointestinal tract (GI) (63.1%, n = 41/65). Infection burden and QoL impact were shown as infections correlated with more severe clinical phenotypes and with a set of relevant non-immune PMM2-CDG signs. Autoimmune diseases had only a marginal presence in PMM2-CDG (2.5%, n = 3/122), all being GI-related. Allergy prevalence was also low in PMM2-CDG (33%, n = 41/122) except for food allergies (26.8%, n = 11/41, of PMM2-CDG and 10.8%, n = 17/158, of controls). High vaccination compliance with greater perceived ineffectiveness (28.3%, n = 17/60) and more severe adverse reactions were described in PMM2-CDG. This people-centric approach not only confirmed literature findings, but created new insights into immunological involvement in CDG, namely by highlighting the possible link between the immune and GI systems in PMM2-CDG. Finally, our results emphasized the importance of patient/caregiver knowledge and raised several red flags about immunological management.
- Published
- 2020
38. Hypothesis: determining phenotypic specificity facilitates understanding of pathophysiology in rare genetic disorders
- Author
-
Peter M. van Hasselt, Hanneke A. Haijes, and Jaak Jaeken
- Subjects
Male ,COG complex ,Glycosylation ,education ,Vesicular Transport Proteins ,Biology ,03 medical and health sciences ,chemistry.chemical_compound ,Congenital Disorders of Glycosylation ,Cog ,Human Phenotype Ontology ,Genetics ,conserved oligomeric Golgi complex ,Humans ,Gene ,health care economics and organizations ,Genetic Association Studies ,Genetics (clinical) ,pathophysiology ,030304 developmental biology ,0303 health sciences ,Conserved oligomeric Golgi complex ,030305 genetics & heredity ,Autophagy ,Hypothesis ,Phenotype ,phenotypic specificity ,humanities ,episodic fever ,Adaptor Proteins, Vesicular Transport ,chemistry ,Multiprotein Complexes ,Episodic fever ,Mutation ,Female ,HPO ,human phenotype ontology - Abstract
In the rapidly growing group of rare genetic disorders, data scarcity demands an intelligible use of available data, in order to improve understanding of underlying pathophysiology. We hypothesize, based on the principle that clinical similarities may be indicative of shared pathophysiology, that determining phenotypic specificity could provide unsuspected insights in pathophysiology of rare genetic disorders. We explored our hypothesis by studying subunit deficiencies of the conserved oligomeric Golgi (COG) complex, a subgroup of congenital disorders of glycosylation (CDG). In this systematic data assessment, all 45 reported patients with COG-CDG were included. The vocabulary of the Human Phenotype Ontology was used to annotate all phenotypic features and to assess occurrence in other genetic disorders. Gene occurrence ratios were calculated by dividing the frequency in the patient cohort over the number of associated genes, according to the Human Phenotype Ontology. Prioritisation based on phenotypic specificity was highly informative and captured phenotypic features commonly associated with glycosylation disorders. Moreover, it captured features not seen in any other glycosylation disorder, among which episodic fever, likely reflecting underappreciated other cellular functions of the COG complex. Interestingly, the COG complex was recently implicated in the autophagy pathway, as are more than half of the genes underlying disorders that present with episodic fever. This suggests that whereas many phenotypic features in these patients are caused by disrupted glycosylation, episodic fever might be caused by disrupted autophagy. Thus, we here demonstrate support for our hypothesis that determining phenotypic specificity could facilitate understanding of pathophysiology in rare genetic disorders. ispartof: JOURNAL OF INHERITED METABOLIC DISEASE vol:43 issue:4 pages:701-711 ispartof: location:United States status: published
- Published
- 2020
39. Congenital disorders of glycosylation: Still 'hot' in 2020
- Author
-
Tomas Honzik, Hana Hansikova, Nina Ondruskova, Jaak Jaeken, and Anna Cechova
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,Glycosylation ,Biophysics ,Disease ,Bioinformatics ,Biochemistry ,Novel gene ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Congenital Disorders of Glycosylation ,Medicine ,Animals ,Humans ,Molecular Biology ,Cellular metabolism ,business.industry ,Disease mechanisms ,Proteins ,Lipid Metabolism ,Lipids ,030104 developmental biology ,chemistry ,Mutation ,business ,Lipid glycosylation ,030217 neurology & neurosurgery ,Metabolic Networks and Pathways - Abstract
Background Congenital disorders of glycosylation (CDG) are inherited metabolic diseases caused by defects in the genes important for the process of protein and lipid glycosylation. With the ever growing number of the known subtypes and discoveries regarding the disease mechanisms and therapy development, it remains a very active field of study. Scope of review This review brings an update on the CDG-related research since 2017, describing the novel gene defects, pathobiomechanisms, biomarkers and the patients' phenotypes. We also summarize the clinical guidelines for the most prevalent disorders and the current therapeutical options for the treatable CDG. Major conclusions In the majority of the 23 new CDG, neurological involvement is associated with other organ disease. Increasingly, different aspects of cellular metabolism (e.g., autophagy) are found to be perturbed in multiple CDG. General significance This work highlights the recent trends in the CDG field and comprehensively overviews the up-to-date clinical recommendations.
- Published
- 2020
40. Aberrant Sialylation in a Patient with a HNF1α Variant and Liver Adenomatosis
- Author
-
Sabrina Giglio, Gert Matthijs, Marie-Cécile Nassogne, Rosangela Artuso, Nicole Revencu, Jaak Jaeken, Gaetano Bertino, Xavier Stéphenne, Domenico Garozzo, Angela Messina, Angelo Palmigiano, Luisa Sturiale, Rita Barone, Immacolata Speciale, and Cristina De Castro
- Subjects
chemistry.chemical_classification ,Glycosylation ,Ms analysis ,Structural diversity ,Biology ,Phenotype ,Sialic acid ,carbohydrates (lipids) ,chemistry.chemical_compound ,chemistry ,Biochemistry ,Wide area ,Transferrin ,Glycoprotein - Abstract
Glycosylation is a fundamental post-translational modification of proteins that boosts their structural diversity providing subtle and specialized biological properties and functions. All those genetic diseases due to a defective glycan biosynthesis and attachment to the nascent glycoproteins fall within the wide area of congenital disorders of glycosylation (CDG), mostly causing multisystem involvement. In the present paper we detailed the unique serum N- glycosylation of a CDG-candidate patient with an unexplained neurological phenotype and liver adenomatosis harbouring a recurrent pathogenic HNF1α variant. Serum transferrin IEF showed a surprising N-glycosylation pattern consisting on hyposialylation as well as remarkable hypersialylation. Mass spectrometry-based glycomic analyses of individual serum glycoproteins enabled to unveil hypersialylated complex N-glycans comprising up to two sialic acids per antenna. Further advanced MS analysis showed the additional sialic acid is bonded through an α2-6 linkage to the peripheral N-acetylglucosamine residue, giving rise to disialo-epitopes never described in human N-glycoproteins.
- Published
- 2020
41. SRD5A3 defective congenital disorder of glycosylation: clinical utility gene card
- Author
-
Jaak Jaeken, Dirk Lefeber, and Gert Matthijs
- Subjects
Metabolic disorders ,Sensitivity and Specificity ,Cataract ,3-Oxo-5-alpha-Steroid 4-Dehydrogenase ,Intellectual Disability ,Genetics ,medicine ,Humans ,Genetic Testing ,Kyphosis ,Gene ,Genetics (clinical) ,Genetic counselling ,business.industry ,Facies ,Membrane Proteins ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,medicine.disease ,Phenotype ,Coloboma ,Mutation (genetic algorithm) ,Mutation ,Clinical Utility Gene Card ,business ,Congenital disorder of glycosylation - Abstract
Contains fulltext : 225786.pdf (Publisher’s version ) (Closed access)
- Published
- 2020
42. Patient and observer reported outcome measures to evaluate health-related quality of life in inherited metabolic diseases: a scoping review
- Author
-
Dorinda Marques-da-Silva, Luísa Barros, Paula A. Videira, Sandra Brasil, Rita Francisco, Vanessa dos Reis Ferreira, Agnes Rafalko, Carlota Pascoal, and Jaak Jaeken
- Subjects
0301 basic medicine ,INBORN-ERRORS ,Health-related quality of life (HrQoL) ,lcsh:Medicine ,Review ,Research & Experimental Medicine ,030105 genetics & heredity ,ENZYME REPLACEMENT THERAPY ,Care setting ,DOUBLE-BLIND ,0302 clinical medicine ,Quality of life ,Surveys and Questionnaires ,Medicine ,Pharmacology (medical) ,ADULT PATIENTS ,Patient reported outcome measures (PROMs) ,FAMILIAL HYPERCHOLESTEROLEMIA ,Genetics (clinical) ,Genetics & Heredity ,FABRY-DISEASE ,AGALSIDASE-BETA ,Outcome measures ,General Medicine ,humanities ,Medicine, Research & Experimental ,Life Sciences & Biomedicine ,medicine.medical_specialty ,Psychometrics ,Pharmacology toxicology ,Inherited metabolic disease(s) (IMD(s)) ,CLINICAL-TRIAL ,03 medical and health sciences ,Metabolic Diseases ,Quality of life (QoL) ,Humans ,TYPE-1 GAUCHER-DISEASE ,Patient Reported Outcome Measures ,ONSET POMPE-DISEASE ,Intensive care medicine ,Observer reported outcome measures (ObsROMs) ,Health related quality of life ,Science & Technology ,business.industry ,lcsh:R ,Clinical research ,Quality of Life ,Observational study ,business ,030217 neurology & neurosurgery ,Social relevance - Abstract
Background Health-related Quality of Life (HrQoL) is a multidimensional measure, which has gained clinical and social relevance. Implementation of a patient-centred approach to both clinical research and care settings, has increased the recognition of patient and/or observer reported outcome measures (PROMs or ObsROMs) as informative and reliable tools for HrQoL assessment. Inherited Metabolic Diseases (IMDs) are a group of heterogeneous conditions with phenotypes ranging from mild to severe and mostly lacking effective therapies. Consequently, HrQoL evaluation is particularly relevant. Objectives We aimed to: (1) identify patient and/or caregiver-reported HrQoL instruments used among IMDs; (2) identify the main results of the application of each HrQoL tool and (3) evaluate the main limitations of HrQoL instruments and study design/methodology in IMDs. Methods A scoping review was conducted using methods outlined by Arksey and O’Malley. Additionally, we critically analysed each article to identify the HrQoL study drawbacks. Results Of the 1954 studies identified, 131 addressed HrQoL of IMDs patients using PROMs and/or ObsROMs, both in observational or interventional studies. In total, we identified 32 HrQoL instruments destined to self- or proxy-completion; only 2% were disease-specific. Multiple tools (both generic and disease-specific) proved to be responsive to changes in HrQoL; the SF-36 and PedsQL questionnaires were the most frequently used in the adult and pediatric populations, respectively. Furthermore, proxy data often demonstrated to be a reliable approach complementing self-reported HrQoL scores. Nevertheless, numerous limitations were identified especially due to the rarity of these conditions. Conclusions HrQoL is still not frequently assessed in IMDs. However, our results show successful examples of the use of patient-reported HrQoL instruments in this field. The importance of HrQoL measurement for clinical research and therapy development, incites to further research in HrQoL PROMs’ and ObsROMs’ creation and validation in IMDs. Electronic supplementary material The online version of this article (10.1186/s13023-018-0953-9) contains supplementary material, which is available to authorized users.
- Published
- 2018
43. Cardiac complications of congenital disorders of glycosylation (CDG): a systematic review of the literature
- Author
-
V. dos Reis Ferreira, Dorinda Marques-da-Silva, D. Webster, Rita Francisco, Thomas Pulinilkunnil, and Jaak Jaeken
- Subjects
0301 basic medicine ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Glycosylation ,Heart Diseases ,Heart disease ,Glycosylphosphatidylinositol ,macromolecular substances ,Biology ,Bioinformatics ,03 medical and health sciences ,chemistry.chemical_compound ,Heart disorder ,Congenital Disorders of Glycosylation ,0302 clinical medicine ,COG complex ,Internal medicine ,Genetics ,medicine ,Animals ,Humans ,Genetics (clinical) ,medicine.disease ,Human genetics ,3. Good health ,carbohydrates (lipids) ,Phenotype ,030104 developmental biology ,Endocrinology ,chemistry ,lipids (amino acids, peptides, and proteins) ,030217 neurology & neurosurgery ,Dolichol synthesis - Abstract
Congenital disorders of glycosylation (CDG) are inborn errors of metabolism due to protein and lipid hypoglycosylation. This rapidly growing family of genetic diseases comprises 103 CDG types, with a broad phenotypic diversity ranging from mild to severe poly-organ -system dysfunction. This literature review summarizes cardiac involvement, reported in 20% of CDG. CDG with cardiac involvement were divided according to the associated type of glycosylation: N-glycosylation, O-glycosylation, dolichol synthesis, glycosylphosphatidylinositol (GPI)-anchor biosynthesis, COG complex, V-ATPase complex, and other glycosylation pathways. The aim of this review was to document and interpret the incidence of heart disease in CDG patients. Heart disorders were grouped into cardiomyopathies, structural defects, and arrhythmogenic disorders. This work may contribute to improved early management of cardiac complications in CDG.
- Published
- 2017
44. Erratum to: What is new in CDG?
- Author
-
Jaak Jaeken and Romain Péanne
- Subjects
0301 basic medicine ,03 medical and health sciences ,030104 developmental biology ,business.industry ,Genetics ,Medicine ,Computational biology ,business ,Genetics (clinical) ,Human genetics - Published
- 2017
45. ORAL D-GALACTOSE SUPPLEMENTATION IN PGM1-CDG
- Author
-
Miao He, Jozef Hertecant, Jolanta Sykut-Cegielska, Nurulamin Abu Bakar, Sunnie Yan Wai Wong, Francis Bowling, David Nguyen, Stefanie Perez, Tim L. Emmerzaal, Katja S. Brocke Holmefjord, Jaak Jaeken, Kea Crivelly, Gernot Poschet, Dieter Koch, Amanda M. Ackermann, Eva Morava, François Foulquier, Dirk Lefeber, Hana Hansikova, Nicole Peeters, Marit Mork, K. Michael Gibson, Kimiyo Raymond, Therese Gadomski, Graeme Preston, Christian Thiel, Monique van Scherpenzeel, Tomas Honzik, Tamas Kozicz, Tulane University, Radboud University Medical Center [Nijmegen], First Faculty of Medicine Charles University [Prague], University of Stavanger, University Hospitals Leuven [Leuven], Washington State University (WSU), Unité de Glycobiologie Structurale et Fonctionnelle UMR 8576 (UGSF), Université de Lille-Centre National de la Recherche Scientifique (CNRS), and Institut National de la Recherche Agronomique (INRA)-Université de Lille-Centre National de la Recherche Scientifique (CNRS)
- Subjects
0301 basic medicine ,Blood Glucose ,Male ,D-galactose ,Stress-related disorders Donders Center for Medical Neuroscience [Radboudumc 13] ,Mannose ,Administration, Oral ,Pilot Projects ,N-glycosylation ,glycomics ,chemistry.chemical_compound ,Prospective Studies ,Child ,Creatine Kinase ,Genetics (clinical) ,Skin ,chemistry.chemical_classification ,O-glycosylation ,biology ,medicine.diagnostic_test ,Transferrin ,Metabolic Disorders Radboud Institute for Molecular Life Sciences [Radboudumc 6] ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Glycogen Storage Disease ,3. Good health ,[CHIM.THEO]Chemical Sciences/Theoretical and/or physical chemistry ,liver function ,Child, Preschool ,Administration ,Female ,Drug ,Partial thromboplastin time ,Oral ,medicine.medical_specialty ,Glycosylation ,endocrine ,LLO ,Adolescent ,Antithrombin III ,Aspartate transaminase ,Neurophysiology ,phosphoglucomutase 1 ,Article ,Dose-Response Relationship ,03 medical and health sciences ,Young Adult ,Internal medicine ,PGM1 ,medicine ,Humans ,coagulation ,Adverse effect ,Preschool ,Blood Coagulation ,Glycoproteins ,Dose-Response Relationship, Drug ,business.industry ,Galactose ,Infant ,carbohydrates (lipids) ,030104 developmental biology ,Endocrinology ,Alanine transaminase ,chemistry ,Phosphoglucomutase ,biology.protein ,Glycoprotein ,transferrin glycoforms ,business - Abstract
Contains fulltext : 181642.pdf (Publisher’s version ) (Closed access) PurposePhosphoglucomutase-1 deficiency is a subtype of congenital disorders of glycosylation (PGM1-CDG). Previous casereports in PGM1-CDG patients receiving oral D-galactose (D-gal) showed clinical improvement. So far no systematic in vitro and clinical studies have assessed safety and benefits of D-gal supplementation. In a prospective pilot study, we evaluated the effects of oral D-gal in nine patients.MethodsD-gal supplementation was increased to 1.5 g/kg/day (maximum 50 g/day) in three increments over 18 weeks. Laboratory studies were performed before and during treatment to monitor safety and effect on serum transferrin-glycosylation, coagulation, and liver and endocrine function. Additionally, the effect of D-gal on cellular glycosylation was characterized in vitro.ResultsEight patients were compliant with D-gal supplementation. No adverse effects were reported. Abnormal baseline results (alanine transaminase, aspartate transaminase, activated partial thromboplastin time) improved or normalized already using 1 g/kg/day D-gal. Antithrombin-III levels and transferrin-glycosylation showed significant improvement, and increase in galactosylation and whole glycan content. In vitro studies before treatment showed N-glycan hyposialylation, altered O-linked glycans, abnormal lipid-linked oligosaccharide profile, and abnormal nucleotide sugars in patient fibroblasts. Most cellular abnormalities improved or normalized following D-gal treatment. D-gal increased both UDP-Glc and UDP-Gal levels and improved lipid-linked oligosaccharide fractions in concert with improved glycosylation in PGM1-CDG.ConclusionOral D-gal supplementation is a safe and effective treatment for PGM1-CDG in this pilot study. Transferrin glycosylation and ATIII levels were useful trial end points. Larger, longer-duration trials are ongoing.
- Published
- 2017
46. MALDI-MS profiling of serumO-glycosylation andN-glycosylation in COG5-CDG
- Author
-
Domenico Garozzo, Luisa Sturiale, Angelo Palmigiano, Daisy Rymen, Rita Barone, Luc Régal, Jaak Jaeken, Cheuk-Wing Fung, Rosaria Ornella Bua, Carlo Dionisi-Vici, and Nicolas Deconinck
- Subjects
0301 basic medicine ,chemistry.chemical_classification ,congenital, hereditary, and neonatal diseases and abnormalities ,Glycan ,Microcephaly ,Glycosylation ,biology ,Glycoconjugate ,Protein subunit ,medicine.disease ,Molecular biology ,carbohydrates (lipids) ,03 medical and health sciences ,chemistry.chemical_compound ,030104 developmental biology ,Cog ,chemistry ,N-linked glycosylation ,Transferrin ,biology.protein ,medicine ,lipids (amino acids, peptides, and proteins) ,Spectroscopy - Abstract
Congenital disorders of glycosylation (CDG) are due to defective glycosylation of glycoconjugates. Conserved oligomeric Golgi (COG)-CDG are genetic diseases due to defects of the COG complex subunits 1-8 causing N-glycan and O-glycan processing abnormalities. In COG-CDG, isoelectric focusing separation of undersialylated glycoforms of serum transferrin and apolipoprotein C-III (apoC-III) allows to detect N-glycosylation and O-glycosylation defects, respectively. COG5-CDG (COG5 subunit deficiency) is a multisystem disease with dysmorphic features, intellectual disability of variable degree, seizures, acquired microcephaly, sensory defects and autistic behavior. We applied matrix-assisted laser desorption/ionization-MS for a high-throughput screening of differential serum O-glycoform and N-glycoform in five patients with COG5-CDG. When compared with age-matched controls, COG5-CDG showed a significant increase of apoC-III0a (aglycosylated glycoform), whereas apoC-III1 (mono-sialylated glycoform) decreased significantly. Serum N-glycome of COG5-CDG patients was characterized by the relative abundance of undersialylated and undergalactosylated biantennary and triantennary glycans as well as slight increase of high-mannose structures and hybrid glycans. Using advanced and well-established MS-based approaches, the present findings reveal novel aspects on O-glycan and N-glycan profiling in COG5-CDG patients, thus providing an increase of current knowledge on glycosylation defects caused by impairment of COG subunits, in support of clinical diagnosis. Copyright © 2017 John Wiley & Sons, Ltd.
- Published
- 2017
47. Further delineation of a rare recessive encephalomyopathy linked to mutations in GFER thanks to data sharing of whole exome sequencing data
- Author
-
C. Thauvin-Robinet, Nada Houcinat, Daphné Lehalle, A Vital, Laurence Faivre, Agnès Rötig, Jaak Jaeken, Fan Xia, Matthew N. Bainbridge, Marlène Rio, Sophie Nambot, Jean-Baptiste Rivière, Paul Kuentz, Julien Thevenon, D Gavrilov, Cyril Goizet, Ange-Line Bruel, Fanny Mochel, N Niu, Arthur L. Beaudet, Yannis Duffourd, Centre de génétique - Centre de référence des maladies rares, anomalies du développement et syndromes malformatifs (CHU de Dijon), Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand ( CHU Dijon ), Lipides - Nutrition - Cancer [Dijon - U1231] ( LNC ), Université de Bourgogne ( UB ) -AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale ( INSERM ), FHU TRANSLAD, Laboratoire de cytogénétique (CHU de Dijon), Centre Hospitalier Universitaire de Dijon - Hôpital François Mitterrand (CHU Dijon), Lipides - Nutrition - Cancer [Dijon - U1231] (LNC), Université de Bourgogne (UB)-AgroSup Dijon - Institut National Supérieur des Sciences Agronomiques, de l'Alimentation et de l'Environnement-Institut National de la Santé et de la Recherche Médicale (INSERM), and FHU TRANSLAD (CHU de Dijon)
- Subjects
Adult ,Male ,0301 basic medicine ,Heterozygote ,Candidate gene ,Adolescent ,data sharing ,Mitochondrial disease ,Compound heterozygosity ,Bioinformatics ,Young Adult ,03 medical and health sciences ,Mitochondrial myopathy ,Mitochondrial Encephalomyopathies ,Exome Sequencing ,Genetics ,Humans ,Medicine ,Genetic Predisposition to Disease ,Oxidoreductases Acting on Sulfur Group Donors ,whole-exome sequencing ,Child ,Exome ,Cytochrome Reductases ,Genetics (clinical) ,Exome sequencing ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,business.industry ,GFER ,Disease gene identification ,medicine.disease ,Pedigree ,3. Good health ,030104 developmental biology ,mitochondrial condition ,Mutation ,Congenital cataracts ,Female ,[ SDV.GEN ] Life Sciences [q-bio]/Genetics ,business - Abstract
Background Alterations in GFER gene have been associated with progressive mitochondrial myopathy, congenital cataracts, hearing loss, developmental delay, lactic acidosis and respiratory chain deficiency in 3 siblings born to consanguineous Moroccan parents by homozygosity mapping and candidate gene approach (OMIM#613076). Next generation sequencing recently confirmed this association by the finding of compound heterozygous variants in 19-year-old girl with a strikingly similar phenotype, but this ultra-rare entity remains however unknown from most of the scientific community. Materials and methods Whole exome sequencing was performed as part of a "diagnostic odyssey" for suspected mitochondrial condition in 2 patients, presenting congenital cataracts, progressive encephalomyopathy and hypotrophy and detected unreported compound heterozygous variants in GFER. Results Thanks to an international data sharing, we found 2 additional patients carrying compound heterozygous variants in GFER. Reverse phenotyping confirmed the phenotypical similarities between the 4 patients. Together with the first literature reports, the review of these 8 cases from 4 unrelated families enables us to better describe this apparently homogeneous disorder, with the clinical and biological stigmata of mitochondrial disease. Conclusion This report highlights the clinical utility of whole exome sequencing and reverse phenotyping for the diagnosis of ultra-rare diseases and underlines the importance of a broad data sharing for accurate clinical delineation of previously unrecognized entities.
- Published
- 2017
48. Galactose Supplementation in Patients With TMEM165-CDG Rescues the Glycosylation Defects
- Author
-
Gert Matthijs, Eva Morava, Leslie K. Climer, Sunnie Wong, Willy Morelle, Vladimir Lupashin, Peter Witters, David Cassiman, Sven Potelle, Therese Gadomski, François Foulquier, Jaak Jaeken, Unité de Glycobiologie Structurale et Fonctionnelle UMR 8576 (UGSF), Université de Lille-Centre National de la Recherche Scientifique (CNRS), University Hospitals Leuven [Leuven], Tulane University, University of Arkansas for Medical Sciences (UAMS), Center for Human Genetics, University of Leuven School of Medicine, SCHOOL of MEDICINE [Louvain], Université Catholique de Louvain = Catholic University of Louvain (UCL)-Université Catholique de Louvain = Catholic University of Louvain (UCL), ANR-15-CE14-0001,SOLV_CDG,Décryptage des patients CDG (Congenital Disorders of Glyvosylation) déficients en TMEM165 - de la compréhension des mécanismes moléculaires à une thérapie(2015), European Project: 643578,H2020,H2020-HCO-2014,E-Rare-3(2014), and Université de Lille-Institut National de la Recherche Agronomique (INRA)-Centre National de la Recherche Scientifique (CNRS)
- Subjects
Male ,0301 basic medicine ,Glycosylation ,Endocrinology, Diabetes and Metabolism ,Clinical Biochemistry ,Biochemistry ,Antiporters ,chemistry.chemical_compound ,Congenital Disorders of Glycosylation ,Endocrinology ,Child ,Cation Transport Proteins ,Membrane Protein ,chemistry.chemical_classification ,biology ,medicine.diagnostic_test ,3. Good health ,[CHIM.THEO]Chemical Sciences/Theoretical and/or physical chemistry ,Treatment Outcome ,lipids (amino acids, peptides, and proteins) ,Adult ,congenital, hereditary, and neonatal diseases and abnormalities ,medicine.medical_specialty ,Glycan ,Context (language use) ,Abnormal glycosylation ,03 medical and health sciences ,Glycolipid ,Western blot ,Internal medicine ,medicine ,Humans ,Clinical Research Articles ,business.industry ,Biochemistry (medical) ,Membrane Proteins ,Galactose ,Fibroblasts ,carbohydrates (lipids) ,HEK293 Cells ,030104 developmental biology ,chemistry ,Transferrin ,Mutation ,Dietary Supplements ,biology.protein ,business - Abstract
International audience; Context: TMEM165 deficiency is a severe multisystem disease that manifests with metabolic, endocrine, and skeletal involvement. It leads to one type of congenital disorders of glycosylation (CDG), a rapidly growing group of inherited diseases in which the glycosylation process is altered. Patients have decreased galactosylation by serum glycan analysis. There are >100 CDGs, but only specific types are treatable. Objective: Galactose has been shown to be beneficial in other CDG types with abnormal galactosylation. The aim of this study was to characterize the effects of galactose supplementation on Golgi glycosylation in TMEM165-depleted HEK293 cells, as well as in 2 patients with TMEM165-CDG and in their cultured skin fibroblast cells. Design and Setting: Glycosylation was assessed by mass spectrometry, western blot analysis, and transferrin isoelectrofocusing. Patients and Interventions: Both unrelated patients with TMEM165-CDG with the same deep intronic homozygous mutation (c.792+182G>A) were allocated to receive d-galactose in a daily dose of 1 g/kg. Results: We analyzed N-linked glycans and glycolipids in knockout TMEM165 HEK293 cells, revealing severe hypogalactosylation and GalNAc transfer defects. Although these defects were completely corrected by the addition of Mn2+, we demonstrated that the observed N-glycosylation defect could also be overcome by galactose supplementation. We then demonstrated that oral galactose supplementation in patients with TMEM165-deficient CDG improved biochemical and clinical parameters, including a substantial increase in the negatively charged transferrin isoforms, and a decrease in hypogalactosylated total N-glycan structures, endocrine function, and coagulation parameters. Conclusion: To our knowledge, this is the first description of abnormal glycosylation of lipids in the TMEM165 defect and the first report of successful dietary treatment in TMEM165 deficiency. We recommend the use of oral d-galactose therapy in TMEM165-CDG.
- Published
- 2017
49. CDG and immune response: From bedside to bench and back
- Author
-
Carlota Pascoal, Vanessa dos Reis Ferreira, Jaak Jaeken, Rita Francisco, Tiago Ferro, and Paula A. Videira
- Subjects
Autoimmune disease ,Inflammation ,congenital, hereditary, and neonatal diseases and abnormalities ,Glycosylation ,business.industry ,NFAT ,medicine.disease ,Abnormal glycosylation ,chemistry.chemical_compound ,Immune system ,Congenital Disorders of Glycosylation ,Phenotype ,chemistry ,Immunity ,Immunology ,Genetics ,medicine ,Humans ,business ,Cell activation ,Genetics (clinical) ,Immunodeficiency - Abstract
Glycosylation is an essential biological process that adds structural and functional diversity to cells and molecules, participating in physiological processes such as immunity. The immune response is driven and modulated by protein-attached glycans that mediate cell-cell interactions, pathogen recognition and cell activation. Therefore, abnormal glycosylation can be associated with deranged immune responses. Within human diseases presenting immunological defects are congenital disorders of glycosylation (CDG), a family of around 130 rare and complex genetic diseases. In this review, we have identified 23 CDG with immunological involvement, characterized by an increased propensity to-often life-threatening-infection. Inflammatory and autoimmune complications were found in 7 CDG types. CDG natural history(ies) and the mechanisms behind the immunological anomalies are still poorly understood. However, in some cases, alterations in pathogen recognition and intracellular signaling (eg, TGF-β1, NFAT, and NF-κB) have been suggested. Targeted therapies to restore immune defects are only available for PGM3-CDG and SLC35C1-CDG. Fostering research on glycoimmunology may elucidate the involved pathophysiological mechanisms and open new therapeutic avenues, thus improving CDG patients' quality of life.
- Published
- 2019
50. NOVEL GLYCOSYLATION INSIGHTS IN ALG12-CDG
- Author
-
Luisa Sturiale, Sebastiano Bianca, Domenico Garozzo, Alessandra Terracciano, Emanuele Agolini, Angela Messina, Angelo Palmigiano, Francesca Esposito, Chiara Barone, Antonio Novelli, Agata Fiumara, Jaak Jaeken, and Rita Barone
- Subjects
congenital, hereditary, and neonatal diseases and abnormalities ,ALG12-CDG ,novel variants ,immunodeficiency ,IgG ,glycophenotype - Abstract
Congenital disorders of glycosylation (CDG) are inherited metabolic diseases affecting the glycan biosynthesis of glycoconjugates [1]. They represent an expanding group of multisystemic diseases with variable phenotypes and prevalent neurological involvement. Among the genetic disorders associated to the protein N-glycosylation pathway, CDG type I (CDG-I) occur in the cytosol or in the endoplasmic reticulum (ER) affecting dolichol-linked oligosaccharide synthesis, whereas CDG type II (CDG-II) involve the N-linked oligosaccharide processing in the Golgi. ALG12-CDG is caused by deficiency of ALG12 ?1,6-mannosyltransferase that adds the eighth mannose residue on the dolichol-PP-oligosaccharide precursor in the ER, thus ensuring its correct shape and branching before the oligosaccharyl-transferase (OST) complex action. We investigated the glyco-phenotype of a novel ALG12-CDG patient with unreported variants, sharing most of the clinical signs of ALG12 deficiency, including recurrent infections and hypogammaglobulinemia. By a combination of consolidated and innovative mass spectrometry-based protocols [2-4], we analyzed serum native transferrin and profiled serum IgG and total serum N-glycans. MALDI MS analysis of intact transferrin showed underoccupancy of N-glycosylation sites, a typical feature of CDG-I. Moreover, N-glycome analysis either on total serum N-glycan pool and on IgG released N-glycans, revealed the abnormal occurrence of high-mannose and hybrid N-glycan species. These accumulating glycoforms, further analyzed by UHPLC-ESI MS, corresponded to unbranched structures with ?1,2-terminal mannose residues, previously identified in serum of patients with MAN1B1-CDG (CDG-II defect) [5]. Glycosylation analyses on the observed ALG12-CDG patient revealed a combination of CDG-I and CDG-II defects, these last associated with abnormal IgG N-glycan profile, consistent with the immunophenotype. As a whole, glycan characterization of target glycoproteins may endorse the molecular defect unraveling the complex clinical phenotype of CDG patients.
- Published
- 2019
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.