70 results on '"Delfien Syx"'
Search Results
2. Analysis of matrisome expression patterns in murine and human dorsal root ganglia
- Author
-
Robin Vroman, Rahel S. Hunter, Matthew J. Wood, Olivia C. Davis, Zoë Malfait, Dale S. George, Dongjun Ren, Diana Tavares-Ferreira, Theodore J. Price, Richard J. Miller, Anne-Marie Malfait, Fransiska Malfait, Rachel E. Miller, and Delfien Syx
- Subjects
matrisome ,dorsal root ganglion ,nociceptor ,cell–cell communication ,extracellular matrix ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
The extracellular matrix (ECM) is a dynamic structure of molecules that can be divided into six different categories and are collectively called the matrisome. The ECM plays pivotal roles in physiological processes in many tissues, including the nervous system. Intriguingly, alterations in ECM molecules/pathways are associated with painful human conditions and murine pain models. Nevertheless, mechanistic insight into the interplay of normal or defective ECM and pain is largely lacking. The goal of this study was to integrate bulk, single-cell, and spatial RNA sequencing (RNAseq) datasets to investigate the expression and cellular origin of matrisome genes in male and female murine and human dorsal root ganglia (DRG). Bulk RNAseq showed that about 65% of all matrisome genes were expressed in both murine and human DRG, with proportionally more core matrisome genes (glycoproteins, collagens, and proteoglycans) expressed compared to matrisome-associated genes (ECM-affiliated genes, ECM regulators, and secreted factors). Single cell RNAseq on male murine DRG revealed the cellular origin of matrisome expression. Core matrisome genes, especially collagens, were expressed by fibroblasts whereas matrisome-associated genes were primarily expressed by neurons. Cell–cell communication network analysis with CellChat software predicted an important role for collagen signaling pathways in connecting vascular cell types and nociceptors in murine tissue, which we confirmed by analysis of spatial transcriptomic data from human DRG. RNAscope in situ hybridization and immunohistochemistry demonstrated expression of collagens in fibroblasts surrounding nociceptors in male and female human DRG. Finally, comparing human neuropathic pain samples with non-pain samples also showed differential expression of matrisome genes produced by both fibroblasts and by nociceptors. This study supports the idea that the DRG matrisome may contribute to neuronal signaling in both mouse and human, and that dysregulation of matrisome genes is associated with neuropathic pain.
- Published
- 2023
- Full Text
- View/download PDF
3. Glycosaminoglycan linkage region of urinary bikunin as a potentially useful biomarker for β3GalT6‐deficient spondylodysplastic Ehlers–Danlos syndrome
- Author
-
Mahnaz Nikpour, Fredrik Noborn, Jonas Nilsson, Tim Van Damme, Olivier Kaye, Delfien Syx, Fransiska Malfait, and Göran Larson
- Subjects
bikunin ,Ehlers–Danlos syndrome ,glycopeptides ,glycoproteomics ,glycosaminoglycan linkage region ,linkeropathies ,Diseases of the endocrine glands. Clinical endocrinology ,RC648-665 ,Genetics ,QH426-470 - Abstract
Abstract The spondylodysplastic type of Ehlers–Danlos syndrome (spEDS) is caused by genetic defects in the B4GALT7 or B3GALT6 genes both deranging the biosynthesis of the glycosaminoglycan linkage region of chondroitin/dermatan sulfate and heparan sulfate proteoglycans. In this study, we have analyzed the linkage regions of urinary chondroitin sulfate proteoglycans of three siblings, diagnosed with spEDS and carrying biallelic pathogenic variants of the B3GALT6 gene. Proteoglycans were digested with trypsin, glycopeptides enriched on anion‐exchange columns, depolymerized with chondroitinase ABC, and analyzed by nLC‐MS/MS. In urine of the unaffected mother, the dominating glycopeptide of bikunin/protein AMBP appeared as only one dominating (99.9%) peak with the canonical tetrasaccharide linkage region modification. In contrast, the samples of the three affected siblings contained two different glycopeptide peaks, corresponding to the canonical tetrasaccharide and to the non‐canonical trisaccharide linkage region modifications in individual ratios of 61/38, 73/27, and 59/41. We propose that the relative distribution of glycosaminoglycan linkage regions of urinary bikunin glycopeptides may serve as a phenotypic biomarker in a diagnostic test but also as a biomarker to follow the effect of future therapies in affected individuals.
- Published
- 2022
- Full Text
- View/download PDF
4. Animal Models of Ehlers–Danlos Syndromes: Phenotype, Pathogenesis, and Translational Potential
- Author
-
Robin Vroman, Anne-Marie Malfait, Rachel E. Miller, Fransiska Malfait, and Delfien Syx
- Subjects
Ehlers–Danlos syndromes ,EDS ,animal models ,mouse ,zebrafish ,Genetics ,QH426-470 - Abstract
The Ehlers–Danlos syndromes (EDS) are a group of heritable connective tissues disorders mainly characterized by skin hyperextensibility, joint hypermobility and generalized tissue fragility. Currently, 14 EDS subtypes each with particular phenotypic features are recognized and are caused by genetic defects in 20 different genes. All of these genes are involved in the biosynthesis and/or fibrillogenesis of collagens at some level. Although great progress has been made in elucidating the molecular basis of different EDS subtypes, the pathogenic mechanisms underlying the observed phenotypes remain poorly understood, and consequentially, adequate treatment and management options for these conditions remain scarce. To date, several animal models, mainly mice and zebrafish, have been described with defects in 14 of the 20 hitherto known EDS-associated genes. These models have been instrumental in discerning the functions and roles of the corresponding proteins during development, maturation and repair and in portraying their roles during collagen biosynthesis and/or fibrillogenesis, for some even before their contribution to an EDS phenotype was elucidated. Additionally, extensive phenotypical characterization of these models has shown that they largely phenocopy their human counterparts, with recapitulation of several clinical hallmarks of the corresponding EDS subtype, including dermatological, cardiovascular, musculoskeletal and ocular features, as well as biomechanical and ultrastructural similarities in tissues. In this narrative review, we provide a comprehensive overview of animal models manifesting phenotypes that mimic EDS with a focus on engineered mouse and zebrafish models, and their relevance in past and future EDS research. Additionally, we briefly discuss domestic animals with naturally occurring EDS phenotypes. Collectively, these animal models have only started to reveal glimpses into the pathophysiological aspects associated with EDS and will undoubtably continue to play critical roles in EDS research due to their tremendous potential for pinpointing (common) signaling pathways, unveiling possible therapeutic targets and providing opportunities for preclinical therapeutic interventions.
- Published
- 2021
- Full Text
- View/download PDF
5. The clinical and mutational spectrum of B3GAT3 linkeropathy: two case reports and literature review
- Author
-
Marlies Colman, Tim Van Damme, Elisabeth Steichen-Gersdorf, Franco Laccone, Sheela Nampoothiri, Delfien Syx, Brecht Guillemyn, Sofie Symoens, and Fransiska Malfait
- Subjects
Connective tissue ,Glycosaminoglycans ,GAG Linkeropathies ,B3GAT3 ,Genotype ,Phenotype ,Medicine - Abstract
Abstract Background Proteoglycans are large and structurally complex macromolecules which can be found in abundancy in the extracellular matrix and on the surface of all animal cells. Mutations in the genes encoding the enzymes responsible for the formation of the tetrasaccharide linker region between the proteoglycan core protein and the glycosaminoglycan side chains lead to a spectrum of severe and overlapping autosomal recessive connective tissue disorders, collectively coined the ‘glycosaminoglycan linkeropathies’. Results We report the clinical findings of two novel patients with a complex linkeropathy due to biallelic mutations in B3GAT3, the gene that encodes glucuronosyltransferase I, which catalyzes the addition of the ultimate saccharide to the linker region. We identified a previously reported c.667G > A missense mutation and an unreported homozygous c.416C > T missense mutation. We also performed a genotype and phenotype-oriented literature overview of all hitherto reported patients harbouring B3GAT3 mutations. A total of 23 patients from 10 families harbouring bi-allelic mutations and one patient with a heterozygeous splice-site mutation in B3GAT3 have been reported. They all display a complex phenotype characterized by consistent presence of skeletal dysplasia (including short stature, kyphosis, scoliosis and deformity of the long bones), facial dysmorphology, and spatulate distal phalanges. More variably present are cardiac defects, joint hypermobility, joint dislocations/contractures and fractures. Seven different B3GAT3 mutations have been reported, and although the number of patients is still limited, some phenotype-genotype correlations start to emerge. The more severe phenotypes seem to have mutations located in the substrate acceptor subdomain of the catalytic domain of the glucuronosyltransferase I protein while more mildly affected phenotypes seem to have mutations in the NTP-sugar donor substrate binding subdomain. Conclusions Loss-of-function mutations in B3GAT3 are associated with a complex connective tissue phenotype characterized by disproportionate short stature, skeletal dysplasia, facial dysmorphism, spatulate distal phalanges and -to a lesser extent- joint contractures, joint hypermobility with dislocations, cardiac defects and bone fragility. Based on the limited number of reported patients, some genotype-phenotype correlations start to emerge.
- Published
- 2019
- Full Text
- View/download PDF
6. Loss of TANGO1 Leads to Absence of Bone Mineralization
- Author
-
Brecht Guillemyn, Sheela Nampoothiri, Delfien Syx, Fransiska Malfait, and Sofie Symoens
- Subjects
COLLAGEN SECRETION ,COLLAGENOPATHY ,COPII VESICLES ,OSTEOGENESIS IMPERFECTA ,TANGO1 ,Orthopedic surgery ,RD701-811 ,Diseases of the musculoskeletal system ,RC925-935 - Abstract
ABSTRACT TANGO1 (transport and Golgi organization‐1 homolog) encodes a transmembrane protein, which is located at endoplasmic reticulum (ER) exit sites where it binds bulky cargo, such as collagens, in the lumen and recruits membranes from the ER‐Golgi intermediate compartment (ERGIC) to create an export route for cargo secretion. Mice lacking Mia3 (murine TANGO1 orthologue) show defective secretion of numerous procollagens and lead to neonatal lethality due to insufficient bone mineralization. Recently, aberrant expression of truncated TANGO1 in humans has been shown to cause a mild‐to‐moderate severe collagenopathy associated with dentinogenesis imperfecta, short stature, skeletal abnormalities, diabetes mellitus, and mild intellectual disability. We now show for the first time that complete loss of TANGO1 results in human embryonic lethality with near‐total bone loss and phenocopies the situation of Mia3−/− mice. Whole‐exome sequencing on genomic DNA (gDNA) of an aborted fetus of Indian descent revealed a homozygous 4‐base pair (4‐bp) deletion in TANGO1 that is heterozygously present in both healthy parents. Parental fibroblast studies showed decreased TANGO1 mRNA expression and protein levels. Type I collagen secretion and extracellular matrix organization were normal, supporting a threshold model for clinical phenotype development. As such, our report broadens the phenotypic and mutational spectrum of TANGO1‐related collagenopathies, and underscores the crucial role of TANGO1 for normal bone development, of which deficiency results in a severe‐to‐lethal form of osteochondrodysplasia. © 2021 American Society for Bone and Mineral Research © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
- Published
- 2021
- Full Text
- View/download PDF
7. Aberrant binding of mutant HSP47 affects posttranslational modification of type I collagen and leads to osteogenesis imperfecta.
- Author
-
Delfien Syx, Yoshihiro Ishikawa, Jan Gebauer, Sergei P Boudko, Brecht Guillemyn, Tim Van Damme, Sanne D'hondt, Sofie Symoens, Sheela Nampoothiri, Douglas B Gould, Ulrich Baumann, Hans Peter Bächinger, and Fransiska Malfait
- Subjects
Genetics ,QH426-470 - Abstract
Heat shock protein 47 (HSP47), encoded by the SERPINH1 gene, is a molecular chaperone essential for correct folding of collagens. We report a homozygous p.(R222S) substitution in HSP47 in a child with severe osteogenesis imperfecta leading to early demise. p.R222 is a highly conserved residue located within the collagen interacting surface of HSP47. Binding assays show a significantly reduced affinity of HSP47-R222S for type I collagen. This altered interaction leads to posttranslational overmodification of type I procollagen produced by dermal fibroblasts, with increased glycosylation and/or hydroxylation of lysine and proline residues as shown by mass spectrometry. Since we also observed a normal intracellular folding and secretion rate of type I procollagen, this overmodification cannot be explained by prolonged exposure of the procollagen molecules to the modifying hydroxyl- and glycosyltransferases, as is commonly observed in other types of OI. We found significant upregulation of several molecular chaperones and enzymes involved in procollagen modification and folding on Western blot and RT-qPCR. In addition, we showed that an imbalance in binding of HSP47-R222S to unfolded type I collagen chains in a gelatin sepharose pulldown assay results in increased binding of other chaperones and modifying enzymes. The elevated expression and binding of this molecular ensemble to type I procollagen suggests a compensatory mechanism for the aberrant binding of HSP47-R222S, eventually leading to overmodification of type I procollagen chains. Together, these results illustrate the importance of HSP47 for proper posttranslational modification and provide insights into the molecular pathomechanisms of the p.(R222S) alteration in HSP47, which leads to a severe OI phenotype.
- Published
- 2021
- Full Text
- View/download PDF
8. b3galt6 Knock-Out Zebrafish Recapitulate β3GalT6-Deficiency Disorders in Human and Reveal a Trisaccharide Proteoglycan Linkage Region
- Author
-
Sarah Delbaere, Adelbert De Clercq, Shuji Mizumoto, Fredrik Noborn, Jan Willem Bek, Lien Alluyn, Charlotte Gistelinck, Delfien Syx, Phil L. Salmon, Paul J. Coucke, Göran Larson, Shuhei Yamada, Andy Willaert, and Fransiska Malfait
- Subjects
b3galt6 ,zebrafish ,trisaccharide linkage region ,proteoglycans ,linkeropathies ,Biology (General) ,QH301-705.5 - Abstract
Proteoglycans are structurally and functionally diverse biomacromolecules found abundantly on cell membranes and in the extracellular matrix. They consist of a core protein linked to glycosaminoglycan chains via a tetrasaccharide linkage region. Here, we show that CRISPR/Cas9-mediated b3galt6 knock-out zebrafish, lacking galactosyltransferase II, which adds the third sugar in the linkage region, largely recapitulate the phenotypic abnormalities seen in human β3GalT6-deficiency disorders. These comprise craniofacial dysmorphism, generalized skeletal dysplasia, skin involvement and indications for muscle hypotonia. In-depth TEM analysis revealed disturbed collagen fibril organization as the most consistent ultrastructural characteristic throughout different affected tissues. Strikingly, despite a strong reduction in glycosaminoglycan content, as demonstrated by anion-exchange HPLC, subsequent LC-MS/MS analysis revealed a small amount of proteoglycans containing a unique linkage region consisting of only three sugars. This implies that formation of glycosaminoglycans with an immature linkage region is possible in a pathogenic context. Our study, therefore unveils a novel rescue mechanism for proteoglycan production in the absence of galactosyltransferase II, hereby opening new avenues for therapeutic intervention.
- Published
- 2020
- Full Text
- View/download PDF
9. Delineation of musculocontractural Ehlers–Danlos Syndrome caused by dermatan sulfate epimerase deficiency
- Author
-
Charlotte K. Lautrup, Keng W. Teik, Ai Unzaki, Shuji Mizumoto, Delfien Syx, Heng H. Sin, Irene K. Nielsen, Sara Markholt, Shuhei Yamada, Fransiska Malfait, Naomichi Matsumoto, Noriko Miyake, and Tomoki Kosho
- Subjects
clinical features ,delineation ,dermatan sulfate ,musculocontractural EDS‐DSE ,Genetics ,QH426-470 - Abstract
Abstract Background Musculocontractural Ehlers–Danlos Syndrome (mcEDS) is a rare connective tissue disorder caused by biallelic loss‐of‐function variants in CHST14 (mcEDS‐CHST14) or DSE (mcEDS‐DSE), both of which result in defective dermatan sulfate biosynthesis. Forty‐one patients with mcEDS‐CHST14 and three patients with mcEDS‐DSE have been described in the literature. Methods Clinical, molecular, and glycobiological findings in three additional patients with mcEDS‐DSE were investigated. Results Three patients from two families shared craniofacial characteristics (hypertelorism, blue sclera, midfacial hypoplasia), skeletal features (pectus and spinal deformities, characteristic finger shapes, progressive talipes deformities), skin features (fine or acrogeria‐like palmar creases), and ocular refractive errors. Homozygous pathogenic variants in DSE were found: c.960T>A/p.Tyr320* in patient 1 and c.996dupT/p.Val333Cysfs*4 in patients 2 and 3. No dermatan sulfate was detected in the urine sample from patient 1, suggesting a complete depletion of DS. Conclusion McEDS‐DSE is a congenital multisystem disorder with progressive symptoms involving craniofacial, skeletal, cutaneous, and cardiovascular systems, similar to the symptoms of mcEDS‐CHST14. However, the burden of symptoms seems lower in patients with mcEDS‐DSE.
- Published
- 2020
- Full Text
- View/download PDF
10. A novel splice variant in the N-propeptide of COL5A1 causes an EDS phenotype with severe kyphoscoliosis and eye involvement.
- Author
-
Sofie Symoens, Fransiska Malfait, Philip Vlummens, Trinh Hermanns-Lê, Delfien Syx, and Anne De Paepe
- Subjects
Medicine ,Science - Abstract
BackgroundThe Ehlers-Danlos Syndrome (EDS) is a heritable connective tissue disorder characterized by hyperextensible skin, joint hypermobility and soft tissue fragility. The classic subtype of EDS is caused by mutations in one of the type V collagen genes (COL5A1 and COL5A2). Most mutations affect the type V collagen helical domain and lead to a diminished or structurally abnormal type V collagen protein. Remarkably, only two mutations were reported to affect the extended, highly conserved N-propeptide domain, which plays an important role in the regulation of the heterotypic collagen fibril diameter. We identified a novel COL5A1 N-propeptide mutation, resulting in an unusual but severe classic EDS phenotype and a remarkable splicing outcome.Methodology/principal findingsWe identified a novel COL5A1 N-propeptide acceptor-splice site mutation (IVS6-2A>G, NM_000093.3_c.925-2A>G) in a patient with cutaneous features of EDS, severe progressive scoliosis and eye involvement. Two mutant transcripts were identified, one with an exon 7 skip and one in which exon 7 and the upstream exon 6 are deleted. Both transcripts are expressed and secreted into the extracellular matrix, where they can participate in and perturb collagen fibrillogenesis, as illustrated by the presence of dermal collagen cauliflowers. Determination of the order of intron removal and computational analysis showed that simultaneous skipping of exons 6 and 7 is due to the combined effect of delayed splicing of intron 7, altered pre-mRNA secondary structure, low splice site strength and possibly disturbed binding of splicing factors.Conclusions/significanceWe report a novel COL5A1 N-propeptide acceptor-splice site mutation in intron 6, which not only affects splicing of the adjacent exon 7, but also causes a splicing error of the upstream exon 6. Our findings add further insights into the COL5A1 splicing order and show for the first time that a single COL5A1 acceptor-splice site mutation can perturb splicing of the upstream exon.
- Published
- 2011
- Full Text
- View/download PDF
11. A tapt1 knock-out zebrafish line with aberrant lens development and impaired vision models human early-onset cataract
- Author
-
Tamara Jarayseh, Brecht Guillemyn, Hanna De Saffel, Jan Willem Bek, Delfien Syx, Sofie Symoens, Yannick Gansemans, Filip Van Nieuwerburgh, Sujatha Jagadeesh, Jayarekha Raja, Fransiska Malfait, Paul J. Coucke, Adelbert De Clercq, and Andy Willaert
- Subjects
Genetics ,Genetics (clinical) - Published
- 2023
- Full Text
- View/download PDF
12. Sensory profiling in classical Ehlers-Danlos syndrome: a case-control study revealing pain characteristics, somatosensory changes, and impaired pain modulation
- Author
-
Marlies Colman, Delfien Syx, Inge de Wandele, Lies Rombaut, Deborah Wille, Zoë Malfait, Mira Meeus, Anne-Marie Malfait, Jessica Van Oosterwijck, and Fransiska Malfait
- Subjects
Article - Abstract
Pain is one of the most important, yet poorly understood complaints in heritable connective tissue disorders (HCTD) caused by monogenic defects in extracellular matrix molecules. This is particularly the case for Ehlers-Danlos syndromes (EDS), paradigm collagen-related disorders. This study aimed to identify the pain signature and somatosensory characteristics in the rare classical type of EDS (cEDS) caused by defects in type V or rarely type I collagen. We used static and dynamic quantitative sensory testing and validated questionnaires in 19 individuals with cEDS and 19 matched controls.Individuals with cEDS reported clinically relevant pain/discomfort (VAS ≥5/10 in 32% for average pain intensity the past month) and worse health -related quality of life. Altered sensory profile was found in the cEDS group with higher (p=0.04) detection thresholds for vibration stimuli at the lower limb indicating hypoesthesia, reduced thermal sensitivity with more (pIn conclusion, Individuals with cEDS report chronic pain and worse health-related quality of life, and present altered somatosensory perception. This study is the first to systematically investigate pain and somatosensory characteristics in a genetically defined HCTD and provides interesting insights on the possible role of the ECM in the development and persistence of pain.
- Published
- 2023
- Full Text
- View/download PDF
13. Clinical and molecular features of 66 patients with musculocontractural Ehlers-Danlos syndrome caused by pathogenic variants in CHST14 (mcEDS- CHST14)
- Author
-
Kosuke Mochida, Anne Slavotinek, Roberto Mendoza-Londono, Parul Jayakar, Kiyoshi Kikkawa, Luis E. Figuera, Andreas R. Janecke, Hiroko Morisaki, Takaya Nakane, Nicol C. Voermans, Delfien Syx, Tetsuyuki Kobayashi, Tomoko Kobayashi, Toshihiro Ohura, Klaas J. Wierenga, Tomomi Yamaguchi, Takayuki Morisaki, Mari Minatogawa, Michihiro Kono, William A. Gahl, Judith D. Ranells, Ai Unzaki, Tomoki Kosho, Cynthia J. Tifft, Yoko Aoki, Masumi Ishikawa, Ohsuke Migita, Akiharu Kubo, Naomichi Matsumoto, Fransiska Malfait, Chiho Tokorodani, Yves Lacassie, Tohru Sonoda, Yvonne Hilhorst-Hofstee, Alessandra Maugeri, Glenda Sobey, Noriko Miyake, Ken Ishikawa, Anupriya Kaur, Hiroshi Kawame, Human genetics, and ACS - Atherosclerosis & ischemic syndromes
- Subjects
Joint hypermobility ,medicine.medical_specialty ,Acrogeria ,business.industry ,human genetics ,medicine.disease ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Dermatology ,Hypotonia ,musculoskeletal diseases ,Ehlers–Danlos syndrome ,Genetics ,medicine ,Joint dislocation ,Craniofacial ,medicine.symptom ,Hypertelorism ,Palmar crease ,business ,Genetics (clinical) - Abstract
BackgroundMusculocontractural Ehlers−Danlos syndrome is caused by biallelic loss-of-function variants in CHST14 (mcEDS-CHST14) or DSE (mcEDS-DSE). Although 48 patients in 33 families with mcEDS-CHST14 have been reported, the spectrum of pathogenic variants, accurate prevalence of various manifestations and detailed natural history have not been systematically investigated.MethodsWe collected detailed and comprehensive clinical and molecular information regarding previously reported and newly identified patients with mcEDS-CHST14 through international collaborations.ResultsSixty-six patients in 48 families (33 males/females; 0–59 years), including 18 newly reported patients, were evaluated. Japanese was the predominant ethnicity (27 families), associated with three recurrent variants. No apparent genotype–phenotype correlation was noted. Specific craniofacial (large fontanelle with delayed closure, downslanting palpebral fissures and hypertelorism), skeletal (characteristic finger morphologies, joint hypermobility, multiple congenital contractures, progressive talipes deformities and recurrent joint dislocation), cutaneous (hyperextensibility, fine/acrogeria-like/wrinkling palmar creases and bruisability) and ocular (refractive errors) features were observed in most patients (>90%). Large subcutaneous haematomas, constipation, cryptorchidism, hypotonia and motor developmental delay were also common (>80%). Median ages at the initial episode of dislocation or large subcutaneous haematoma were both 6 years. Nine patients died; their median age was 12 years. Several features, including joint and skin characteristics (hypermobility/extensibility and fragility), were significantly more frequent in patients with mcEDS-CHST14 than in eight reported patients with mcEDS-DSE.ConclusionThis first international collaborative study of mcEDS-CHST14 demonstrated that the subtype represents a multisystem disorder with unique set of clinical phenotypes consisting of multiple malformations and progressive fragility-related manifestations; these require lifelong, multidisciplinary healthcare approaches.
- Published
- 2022
- Full Text
- View/download PDF
14. Caffey disease is associated with distinct arginine to cysteine substitutions in the proα1(I) chain of type I procollagen
- Author
-
Delfien Syx, Geert Mortier, Sofie Symoens, Peter H. Byers, Trinh Hermanns-Lê, Ingrid Hausser, Tibbe Dhooge, Jonathan Zonana, and Fransiska Malfait
- Subjects
Arginine ,Infantile cortical hyperostosis ,Inflammation ,Biology ,medicine.disease ,Molecular biology ,Collagen Type I ,Hyperostosis, Cortical, Congenital ,Collagen Type I, alpha 1 Chain ,Procollagen peptidase ,medicine.anatomical_structure ,Dermis ,Ehlers–Danlos syndrome ,Child, Preschool ,Mutation ,medicine ,Humans ,Allelic heterogeneity ,Cysteine ,Human medicine ,medicine.symptom ,Procollagen ,Genetics (clinical) - Abstract
Purpose Infantile Caffey disease is a rare disorder characterized by acute inflammation with subperiosteal new bone formation, associated with fever, pain, and swelling of the overlying soft tissue. Symptoms arise within the first weeks after birth and spontaneously resolve before the age of two years. Many, but not all, affected individuals carry the heterozygous pathogenic COL1A1 variant (c.3040C>T, p.(Arg1014Cys)). Methods We sequenced COL1A1 in 28 families with a suspicion of Caffey disease and performed ultrastructural, immunocytochemical, and biochemical collagen studies on patient skin biopsies. Results We identified the p.(Arg1014Cys) variant in 23 families and discovered a novel heterozygous pathogenic COL1A1 variant (c.2752C>T, p.(Arg918Cys)) in five. Both arginine to cysteine substitutions are located in the triple helical domain of the pro alpha 1(I) procollagen chain. Dermal fibroblasts (one patient with p.(Arg1014Cys) and one with p.(Arg918Cys)) produced molecules with disulfide-linked pro alpha 1(I) chains, which were secreted only with p.(Arg1014Cys). No intracellular accumulation of type I procollagen was detected. The dermis revealed mild ultrastructural abnormalities in collagen fibril diameter and packing. Conclusion The discovery of this novel pathogenic variant expands the limited spectrum of arginine to cysteine substitutions in type I procollagen. Furthermore, it confirms allelic heterogeneity in Caffey disease and impacts its molecular confirmation.
- Published
- 2021
- Full Text
- View/download PDF
15. The matrisome of the murine and human dorsal root ganglion: a transcriptomal approach
- Author
-
Robin Vroman, Rahel Hunter, Matthew J. Wood, Olivia C. Davis, Zoë Malfait, Dale S. George, Dongjun Ren, Diana Tavares-Ferreira, Theodore J. Price, Anne-Marie Malfait, Fransiska Malfait, Rachel E. Miller, and Delfien Syx
- Abstract
The extracellular matrix (ECM) is a dynamic structure composed of a large number of molecules that can be divided into six different categories and are collectively called the matrisome. The ECM plays pivotal roles in physiological and pathological processes in many tissues, including the nervous system. Intriguingly, alterations in ECM molecules/pathways are often associated with painful human conditions and murine experimental pain models. Nevertheless, mechanistic insight into the interplay of normal or defective ECM and pain is largely lacking. To expand the knowledge on ECM composition and synthesis in the peripheral nervous system, we used a transcriptomal approach to investigate the expression and cellular origin of matrisome genes in murine and human dorsal root ganglia (DRG), containing the cell bodies of sensory neurons. Bulk RNA sequencing data showed that over 60% of all matrisome genes were expressed in both murine and human DRG, with proportionally more core matrisome genes (glycoproteins, collagens, and proteoglycans) expressed compared to matrisome-associated genes (ECM-affiliated genes, ECM regulators and secreted factors). Examination of the cellular origin of matrisome expression by single cell RNA sequencing on murine DRG revealed that core matrisome genes, especially collagens, were expressed by vascular leptomeningeal-like (fibroblast) cell types whereas matrisome-associated genes were mainly expressed by neuronal cell types. We analyzed cell-cell communication networks with the CellChat R package and predicted an important role for the Collagen signaling pathway in connecting vascular cell types and nociceptors in murine tissue, which we confirmed by analysis of spatial transcriptomic data from human DRG. RNAscopein situhybridization and immunohistochemistry confirmed expression of collagens in fibroblasts surrounding nociceptors in human DRG. This study supports the idea that the DRG matrisome may contribute to neuronal signaling in both mouse and human. The identification of the cellular distribution of murine and human matrisome genes provides a framework to study the role of the ECM in peripheral nervous tissue and its effects on pain signaling.HighlightsTranscriptomal analyses of mouse and human dorsal root ganglia (DRG) revealed that over 60% of matrisome genes are expressed by murine and human dorsal root ganglia (DRG), with over 85% of the genes with orthologues overlapping between both species.Matrisome-associated genes had the highest expression in both species and included conserved expression of annexins, S100 calcium binding proteins and cathepsins.Collagens and collagen receptors are expressed by distinct cell types in murine and human DRG, suggesting that the collagen signaling pathway could be involved in cell-cell signaling.
- Published
- 2022
- Full Text
- View/download PDF
16. Alterations in glycosaminoglycan biosynthesis associated with the Ehlers-Danlos syndromes
- Author
-
Delfien Syx, Sarah Delbaere, Catherine Bui, Adelbert De Clercq, Göran Larson, Shuji Mizumoto, Tomoki Kosho, Sylvie Fournel-Gigleux, and Fransiska Malfait
- Subjects
decorin ,collagen ,proteoglycan ,Physiology ,Biology and Life Sciences ,Dermatan Sulfate ,Cell Biology ,Ehlers-Danlos syndromes ,Medicine and Health Sciences ,glycosaminoglycan ,Animals ,Ehlers-Danlos Syndrome ,Proteoglycans ,Collagen ,Sulfotransferases - Abstract
Proteoglycans consist of a core protein substituted with one or more glycosaminoglycan (GAG) chains and execute versatile functions during many physiological and pathological processes. The biosynthesis of GAG chains is a complex process that depends on the concerted action of a variety of enzymes. Central to the biosynthesis of heparan sulfate (HS) and chondroitin sulfate/dermatan sulfate (CS/DS) GAG chains is the formation of a tetrasaccharide linker region followed by biosynthesis of HS or CS/DS-specific repeating disaccharide units, which then undergo modifications and epimerization. The importance of these biosynthetic enzymes is illustrated by several severe pleiotropic disorders that arise upon their deficiency. The Ehlers-Danlos syndromes (EDS) constitute a special group among these disorders. Although most EDS types are caused by defects in fibrillar types I, III, or V collagen, or their modifying enzymes, a few rare EDS types have recently been linked to defects in GAG biosynthesis. Spondylodysplastic EDS (spEDS) is caused by defective formation of the tetrasaccharide linker region, either due to β4GalT7 or β3GalT6 deficiency, whereas musculocontractural EDS (mcEDS) results from deficiency of D4ST1 or DS-epi1, impairing DS formation. This narrative review highlights the consequences of GAG deficiency in these specific EDS types, summarizes the associated phenotypic features and the molecular spectrum of reported pathogenic variants, and defines the current knowledge on the underlying pathophysiological mechanisms based on studies in patient-derived material, in vitro analyses, and animal models.
- Published
- 2022
17. Clinical and molecular characteristics of 168 probands and 65 relatives with a clinical presentation of classical Ehlers–Danlos syndrome
- Author
-
Inge De Wandele, Fransiska Malfait, Delfien Syx, Sofie Symoens, Tibbe Dhooge, and Marlies Colman
- Subjects
Joint Instability ,Proband ,Joint hypermobility ,medicine.medical_specialty ,Genetic counseling ,Carboxypeptidases ,Biology ,medicine.disease ,Dermatology ,Type V collagen ,Repressor Proteins ,Phenotype ,Unknown Significance ,Ehlers–Danlos syndrome ,Mutation ,Genetics ,medicine ,Humans ,Ehlers-Danlos Syndrome ,Presentation (obstetrics) ,Heritable connective tissue disorder ,Collagen Type V ,Genetics (clinical) - Abstract
Classical Ehlers-Danlos syndrome (cEDS) is a heritable connective tissue disorder mainly caused by pathogenic variants in COL5A1 or COL5A2, encoding type V collagen. Its diagnosis, based on clinical criteria and molecular confirmation, can be challenging. We report the molecular and clinical characteristics of 168 probands (72 clinically evaluated at our center) and 65 relatives with a clinical presentation of cEDS. Type V collagen defects were found in 145 probands, 121 (83.5%) were located in COL5A1 and 24 (16.5%) in COL5A2. Although 85.6% of molecularly confirmed patients presented the two major clinical criteria (generalized joint hypermobility, hyperextensible skin with atrophic scarring), significant inter- and intrafamilial phenotypic variability was noted. COL5A2 variants often caused a more severe phenotype. Vascular complications were rare in individuals with type V collagen defects (1.4%). Among the 72 probands clinically evaluated in our center, the mutation detection rate was 82.0%. The majority (68.1%) harbored COL5A1/COL5A2 defects. Yet, 13.9% harbored a defect in another gene (COL1A1, PLOD1, TNXB, AEBP1) highlighting important clinical overlap and the need for molecular confirmation of the diagnosis as this has implications regarding follow-up and genetic counseling. Eighteen percent of the 72 probands remained molecularly unexplained and a COL5A1 variant of unknown significance was identified in 6.9%.
- Published
- 2021
- Full Text
- View/download PDF
18. Pain-related behaviors and abnormal cutaneous innervation in a murine model of classical Ehlers–Danlos syndrome
- Author
-
Robin Vroman, Delfien Syx, A. Obeidat, Fransiska Malfait, Anne-Marie Malfait, P.B. Tran, Richard J. Miller, Rachel E. Miller, and Zoë Malfait
- Subjects
Male ,Nervous system ,medicine.medical_specialty ,Pain ,Nerve fiber ,Article ,Mice ,03 medical and health sciences ,0302 clinical medicine ,030202 anesthesiology ,Internal medicine ,medicine ,Animals ,Sensitization ,Skin ,business.industry ,Chronic pain ,medicine.disease ,Disease Models, Animal ,Anesthesiology and Pain Medicine ,medicine.anatomical_structure ,Endocrinology ,Neurology ,Ehlers–Danlos syndrome ,Mutation ,Nociceptor ,Ehlers-Danlos Syndrome ,Female ,Neurology (clinical) ,business ,Cutaneous innervation ,Haploinsufficiency ,Collagen Type V ,030217 neurology & neurosurgery - Abstract
Classical Ehlers-Danlos syndrome (cEDS) is a connective tissue disorder caused by heterozygous mutations in one of the type V collagen-encoding genes, COL5A1 or COL5A2. cEDS is characterized by generalized joint hypermobility and instability, hyperextensible, fragile skin and delayed wound healing. Chronic pain is a major problem in cEDS patients, but the underlying mechanisms are largely unknown, and studies in animal models are lacking. Therefore, we assessed pain-related behaviors in haploinsufficient Col5a1(+/−) mice, which clinically mimic human cEDS. Compared to wild-type (WT) littermates, 15–20 week-old Col5a1(+/−) mice of both sexes showed significant hypersensitivity to mechanical stimuli in the hind paws and the abdominal area, but responses to thermal stimuli were unaltered. Spontaneous behaviors, including distance travelled and rearing, were grossly normal in male Col5a1(+/−) mice, while female Col5a1(+/−) mice showed altered climbing behavior. Finally, male and female Col5a1(+/−) mice vocalized more than WT littermates when scruffed. Decreased grip strength was also noted. In view of the observed pain phenotype Col5a1(+/−) mice were crossed with Na(V)1.8-tdTomato reporter mice, enabling visualization of nociceptors in the glabrous skin of the footpad. We observed a significant decrease in intra-epidermal nerve fiber density, with fewer nerves crossing the epidermis, and a decreased total nerve length of Col5a1(+/−) mice compared to WT. In summary, male and female Col5a1(+/−) mice show hypersensitivity to mechanical stimuli, indicative of generalized sensitization of the nervous system, in conjunction with an aberrant organization of cutaneous nociceptors. Therefore, Col5a1(+/−) mice will provide a useful tool to study mechanisms of pain associated with cEDS.
- Published
- 2020
- Full Text
- View/download PDF
19. Microarray analyses of the dorsal root ganglia support a role for innate neuro-immune pathways in persistent pain in experimental osteoarthritis
- Author
-
Anne-Marie Malfait, Rachel E. Miller, Delfien Syx, Dongjun Ren, Ana M. Valdes, S. Ishihara, Richard J. Miller, and P.B. Tran
- Subjects
Male ,0301 basic medicine ,CCR2 ,Microarray ,Neuroimmunomodulation ,Biomedical Engineering ,Gene Expression ,Disease ,Osteoarthritis ,Bioinformatics ,Menisci, Tibial ,Article ,Transcriptome ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,Ganglia, Spinal ,Animals ,Medicine ,Orthopedics and Sports Medicine ,RNA, Messenger ,Neuroinflammation ,030203 arthritis & rheumatology ,Pain, Postoperative ,business.industry ,Gene Expression Profiling ,Sham surgery ,Osteoarthritis, Knee ,Microarray Analysis ,medicine.disease ,Arthralgia ,Arthritis, Experimental ,Immunity, Innate ,Phenotype ,030104 developmental biology ,Neuropathic pain ,Disease Progression ,business - Abstract
OBJECTIVE: Following destabilization of the medial meniscus (DMM), mice develop experimental osteoarthritis (OA) and associated pain behaviors that are dependent on the stage of disease. We aimed to describe changes in gene expression in knee-innervating dorsal root ganglia (DRG) after surgery, in order to identify molecular pathways associated with three pre-defined pain phenotypes: “post-surgical pain”, “early-stage OA pain”, and “persistent OA pain”. DESIGN: We performed DMM or sham surgery in 10-week old male C57BL/6 mice and harvested L3-L5 DRG 4, 8, and 16 weeks after surgery or from age-matched naïve mice (n=3/group). RNA was extracted and an Affymetrix Mouse Transcriptome Array 1.0 was performed. Three pain phenotypes were defined: “post-surgical pain” (sham and DMM 4-week vs. 14-week old naïve), “early OA pain” (DMM 4-week vs. sham 4-week), and “persistent OA pain” (DMM 8- and 16-week vs. naïve and sham 8- and 16-week). ‘Top hit’ genes were defined as p
- Published
- 2020
- Full Text
- View/download PDF
20. Exploring pain mechanisms in hypermobile Ehlers-Danlos syndrome: A case-control study
- Author
-
Inge De Wandele, Marlies Colman, Linda Hermans, Jessica Van Oosterwijck, Mira Meeus, Lies Rombaut, Griet Brusselmans, Delfien Syx, Patrick Calders, and Fransiska Malfait
- Subjects
Pain Threshold ,Anesthesiology and Pain Medicine ,Case-Control Studies ,Humans ,Ehlers-Danlos Syndrome ,Female ,Chronic Pain - Abstract
The hypermobile type of Ehlers-Danlos syndrome (hEDS) is a heritable connective tissue disorder, associated with joint hypermobility and prominent chronic pain. Because experimental pain testing in hEDS is scarce, the underlying mechanisms are still poorly understood.The present study assesses endogenous pain facilitation and pain inhibition in hEDS, using a protocol for temporal summation of pain (TSP), conditioned pain modulation (CPM) and exercise-induced hypoalgesia (EIH).Twenty women with hEDS and 20 age-matched healthy controls participated. After evaluating thermal and mechanical pain thresholds (PPT), TSP was assessed using 10 repetitive painful pressure stimuli. CPM was provoked using pressure as the test stimulus and hand immersion in hot water (46°) as the conditioning stimulus. EIH was assessed after a submaximal cycling protocol.The hEDS group demonstrated reduced PPTs and showed significantly more TSP after repeated painful stimuli than the control group. In comparison to the healthy control group, the hEDS group demonstrated significantly less EIH at the quadriceps test location. At the trapezius, EIH did not significantly differ between groups. No significant differences were found between the hEDS group and control group in the CPM response.The results demonstrate increased TSP in hEDS, suggesting increased central pain facilitation. EIH should be studied more extensively but may be disturbed when evaluated in the muscles that are activated during exercise. The CPM results are inconclusive and require more research.Studies regarding the mechanisms that underlie pain in hEDS are scarce, although it is the most prevalent and disabling symptom in this patient population. This study demonstrates increased temporal summation in hEDS and suggests that exercise-induced hypoalgesia may be reduced. Because exercise is a cornerstone in the multidisciplinary treatment of heritable connective tissue disorders, gaining knowledge in this field is important. Pressure stimuli were used to facilitate the international usability of the protocols, allowing for future data acquisition in large cohorts.
- Published
- 2022
21. NRF2 Shortage in Human Skin Fibroblasts Dysregulates Matrisome Gene Expression and Affects Collagen Fibrillogenesis
- Author
-
Mélanie Salamito, Benjamin Gillet, Delfien Syx, Elisabeth Vaganay, Marilyne Malbouyres, Catherine Cerutti, Nicolas Tissot, Chloé Exbrayat-Héritier, Philippe Perez, Christophe Jones, Sandrine Hughes, Fransiska Malfait, Valérie Haydont, Sibylle Jäger, Florence Ruggiero, Institut de Génomique Fonctionnelle de Lyon (IGFL), École normale supérieure de Lyon (ENS de Lyon)-Université Claude Bernard Lyon 1 (UCBL), Université de Lyon-Université de Lyon-Centre National de la Recherche Scientifique (CNRS)-Institut National de Recherche pour l’Agriculture, l’Alimentation et l’Environnement (INRAE), Centre National de la Recherche Scientifique (CNRS), Ghent University Hospital, Center for Medical Genetics [Ghent], L'OREAL, and Research & Innovation
- Subjects
[SDV]Life Sciences [q-bio] ,[SDV.BBM.GTP]Life Sciences [q-bio]/Biochemistry, Molecular Biology/Genomics [q-bio.GN] ,[SDV.BC]Life Sciences [q-bio]/Cellular Biology ,Cell Biology ,Dermatology ,Molecular Biology ,Biochemistry - Abstract
International audience; NRF2 is a master regulator of anti-oxidative response that was recently proposed as a potential regulator of extracellular matrix (ECM) gene expression. Fibroblasts are major ECM producers in all connective tissues including dermis. A better understanding of NRF2-mediated ECM regulation in skin fibroblasts is thus of great interest for skin homeostasis maintenance and aging protection. Here, we investigate the impact of NRF2 downregulation on matrisome gene expression and ECM deposits in human primary dermal fibroblasts. RNA-seq-based transcriptome analysis of NRF2 silenced dermal fibroblasts shows that ECM genes are the most regulated gene sets, highlighting the relevance of the NRF2-mediated matrisome program in these cells. Using complementary light and electron microscopy methods, we show that NRF2 deprivation in dermal fibroblasts results in reduced collagen I biosynthesis and impacts collagen fibril deposition. Moreover, we identify ZNF469, a putative transcriptional regulator of collagen biosynthesis, as a novel target of NRF2. Both ZNF469 silenced fibroblasts and fibroblasts derived from Brittle Corneal Syndrome patients carrying mutations in ZNF469 show reduced collagen I gene expression. Our study shows that NRF2 orchestrates matrisome expression in human skin fibroblasts through direct or indirect transcriptional mechanisms that could be prioritized to target dermal ECM homeostasis in health and disease.
- Published
- 2023
- Full Text
- View/download PDF
22. Atypical variants in COL1A1 and COL3A1 associated with classical and vascular Ehlers-Danlos syndrome overlap phenotypes: expanding the clinical phenotype based on additional case reports
- Author
-
Marlies Colman, Marco Castori, Lucia Micale, Marco Ritelli, Marina Colombi, Neeti Ghali, Fleur Van Dijk, Luisa Marsili, Adrienne Weeks, Anthony Vandersteen, Andrea Rideout, Anne Legrand, Michael Frank, Tristan Mirault, Alessandro Ferraris, Niccolò Di Giosaffatte, Paola Grammatico, Juergen Grunert, Charissa Frank, Sofie Symoens, Delfien Syx, and Fransiska Malfait
- Subjects
vascular rupture ,Immunology ,skin hyperextensibility ,joint hypermobility ,Collagen Type I, alpha 1 Chain ,Collagen Type III ,Phenotype ,Rheumatology ,Mutation ,Immunology and Allergy ,Humans ,Ehlers-Danlos Syndrome ,Collagen ,Ehlers-Danlos syndrome ,collagen ,connective tissue - Abstract
The vast majority of reported (likely) pathogenic missense variants in the genes coding for the fibrillar collagens leads to the substitution of one of the obligatory glycine residues in the Gly-Xaa-Yaa repeat sequence of the triple helical domain. Their phenotypic consequences and deleterious effects have been well-documented. However, with increasing access to molecular diagnostic testing based on next-generation sequencing techniques, such as sequencing of multi-gene panels and whole-exome sequencing, non-glycine substitutions are more frequently identified in individuals suspected to have a heritable collagen disorder, but their pathogenic effect is often difficult to predict.Some specific non-glycine substitutions in the proα1(I)- (p.(Arg312Cys)) and proα1(III)- (glutamic acid to lysine at different positions) collagen chain have been identified in a number of individuals presenting a phenotype showing features of both classical and vascular Ehlers-Danlos syndrome. The number of reported individuals with these defects is currently very low, and several of these non-glycine substitutions had initially been categorised as variants of unknown significance (VUS), complicating early diagnosis, accurate counselling, management guidelines, and correct classification. This collaborative study reports on the phenotype of 22 and 7 individuals harbouring these rare variants in COL1A1 and COL3A1, respectively, expanding our knowledge on clinical presentation, phenotypic variability, and natural history, and informing on the risk for potentially life-threatening events, such as vascular, gastro-intestinal, and pregnancy-related complications.
- Published
- 2021
23. Pain in the Ehlers-Danlos syndromes: Mechanisms, models, and challenges
- Author
-
Delfien Syx, Anne-Marie Malfait, Fransiska Malfait, Lies Rombaut, Rachel E. Miller, Robin Vroman, Marlies Colman, and Inge De Wandele
- Subjects
Joint hypermobility ,Joint Instability ,medicine.medical_specialty ,Heterogeneous group ,business.industry ,Chronic pain ,Connective tissue ,Pain ,medicine.disease ,Ehlers danlos ,Collagen biosynthesis ,medicine.anatomical_structure ,Genetics ,Skin Abnormalities ,Medicine ,Humans ,Skin hyperextensibility ,Ehlers-Danlos Syndrome ,business ,Intensive care medicine ,Connective Tissue Diseases ,Psychosocial ,Genetics (clinical) - Abstract
Chronic pain is one of the most common, yet poorly studied, complaints in people suffering from Ehlers-Danlos syndromes (EDS). This heterogeneous group of heritable connective tissue disorders is typically characterized by skin hyperextensibility, joint hypermobility, and generalized connective tissue fragility. Most EDS types are caused by genetic defects that affect connective tissue biosynthesis, thereby compromising collagen biosynthesis or fibrillogenesis and resulting in a disorganized extracellular matrix. Even though chronic pain is a major source of disability, functional impairment, and psychosocial suffering in EDS, currently used analgesics and other treatment strategies provide inadequate pain relief and thus represents an important unmet medical need. An important contributor to this is the lack of knowledge about the underlying mechanisms. In this narrative review, we summarize the current understanding of pain and the associated mechanisms in EDS based on clinical studies focusing on questionnaires and experimental pain testing as well as studies in animal models of EDS. In addition, we highlight the challenges, gaps, and opportunities in EDS-pain research.
- Published
- 2021
24. Animal Models of Ehlers–Danlos Syndromes: Phenotype, Pathogenesis, and Translational Potential
- Author
-
Delfien Syx, Anne-Marie Malfait, Rachel E. Miller, Robin Vroman, and Fransiska Malfait
- Subjects
Review ,CLINICAL VARIABILITY ,TARGETED DISRUPTION ,Biology ,QH426-470 ,DEFICIENT MICE ,EDS ,Pathogenesis ,MISSENSE MUTATION ,MATRIX TENASCIN-X ,Ehlers–Danlos syndromes ,Medicine and Health Sciences ,Genetics ,Genetics(clinical) ,Skin hyperextensibility ,Zebrafish ,Genetics (clinical) ,mouse ,V COLLAGEN ,Phenocopy ,Biology and Life Sciences ,MURINE MODEL ,Fibrillogenesis ,biology.organism_classification ,zebrafish ,Phenotype ,animal models ,Ehlers danlos ,Collagen biosynthesis ,CONNECTIVE-TISSUE ,III COLLAGEN ,CARBOXYPEPTIDASE-LIKE PROTEIN ,Ehlers-Danlos syndromes ,Molecular Medicine ,Neuroscience - Abstract
The Ehlers–Danlos syndromes (EDS) are a group of heritable connective tissues disorders mainly characterized by skin hyperextensibility, joint hypermobility and generalized tissue fragility. Currently, 14 EDS subtypes each with particular phenotypic features are recognized and are caused by genetic defects in 20 different genes. All of these genes are involved in the biosynthesis and/or fibrillogenesis of collagens at some level. Although great progress has been made in elucidating the molecular basis of different EDS subtypes, the pathogenic mechanisms underlying the observed phenotypes remain poorly understood, and consequentially, adequate treatment and management options for these conditions remain scarce. To date, several animal models, mainly mice and zebrafish, have been described with defects in 14 of the 20 hitherto known EDS-associated genes. These models have been instrumental in discerning the functions and roles of the corresponding proteins during development, maturation and repair and in portraying their roles during collagen biosynthesis and/or fibrillogenesis, for some even before their contribution to an EDS phenotype was elucidated. Additionally, extensive phenotypical characterization of these models has shown that they largely phenocopy their human counterparts, with recapitulation of several clinical hallmarks of the corresponding EDS subtype, including dermatological, cardiovascular, musculoskeletal and ocular features, as well as biomechanical and ultrastructural similarities in tissues. In this narrative review, we provide a comprehensive overview of animal models manifesting phenotypes that mimic EDS with a focus on engineered mouse and zebrafish models, and their relevance in past and future EDS research. Additionally, we briefly discuss domestic animals with naturally occurring EDS phenotypes. Collectively, these animal models have only started to reveal glimpses into the pathophysiological aspects associated with EDS and will undoubtably continue to play critical roles in EDS research due to their tremendous potential for pinpointing (common) signaling pathways, unveiling possible therapeutic targets and providing opportunities for preclinical therapeutic interventions.
- Published
- 2021
- Full Text
- View/download PDF
25. Aberrant binding of mutant HSP47 affects posttranslational modification of type I collagen and leads to osteogenesis imperfecta
- Author
-
Brecht Guillemyn, Fransiska Malfait, Sofie Symoens, Jan M. Gebauer, Sheela Nampoothiri, Tim Van Damme, Hans Peter Bächinger, Douglas B. Gould, Yoshihiro Ishikawa, Sergei P. Boudko, Sanne D'hondt, Ulrich Baumann, Delfien Syx, and Bateman, John F
- Subjects
TRIPLE-HELIX ,Mutant ,Sequence Homology ,Plasma protein binding ,Biochemistry ,Heat Shock Response ,Binding Analysis ,Models ,Animal Cells ,Child ,Cells, Cultured ,Connective Tissue Cells ,Heat shock protein 47 ,0303 health sciences ,Cultured ,030305 genetics & heredity ,Cell biology ,Child, Preschool ,Cellular Types ,Type I collagen ,Glycosylation ,Cells ,1.1 Normal biological development and functioning ,ENDOPLASMIC-RETICULUM ,FKBP65 ,03 medical and health sciences ,Rheumatology ,Protein Domains ,Genetics ,Humans ,Amino Acid Sequence ,Molecular Biology Techniques ,Molecular Biology ,Secretion ,Chemical Characterization ,Ecology, Evolution, Behavior and Systematics ,Molecular Biology Assays and Analysis Techniques ,Post-Translational ,RECOGNITION ,Molecular ,Biology and Life Sciences ,Proteins ,Infant ,Fibroblasts ,Procollagen peptidase ,Biological Tissue ,chemistry ,SHOCK-PROTEIN 47 ,Mutation ,Physiological Processes ,Developmental Biology ,Models, Molecular ,Cancer Research ,Physiology ,VARIANT ,Lysine ,QH426-470 ,chemistry.chemical_compound ,Fatal Outcome ,Animal Products ,Medicine and Health Sciences ,PROPEPTIDE ,Connective Tissue Diseases ,Genetics (clinical) ,Cellular Stress Responses ,Pediatric ,MOLECULAR CHAPERONE ,biology ,Agriculture ,Osteogenesis Imperfecta ,Recombinant Proteins ,DEFICIENCY ,Amino Acid ,CHAPERONE HSP47 ,Connective Tissue ,Cell Processes ,embryonic structures ,Amino Acid Analysis ,Female ,Anatomy ,Research Article ,Protein Binding ,animal structures ,Mutation, Missense ,macromolecular substances ,Research and Analysis Methods ,Collagen Type I ,Rare Diseases ,Underpinning research ,Preschool ,Protein precursor ,HSP47 Heat-Shock Proteins ,Protein Processing ,030304 developmental biology ,Sequence Homology, Amino Acid ,Collagen Diseases ,Infant, Newborn ,Cell Biology ,Newborn ,biology.protein ,Gelatin ,Missense ,Collagens ,Protein Processing, Post-Translational - Abstract
Heat shock protein 47 (HSP47), encoded by the SERPINH1 gene, is a molecular chaperone essential for correct folding of collagens. We report a homozygous p.(R222S) substitution in HSP47 in a child with severe osteogenesis imperfecta leading to early demise. p.R222 is a highly conserved residue located within the collagen interacting surface of HSP47. Binding assays show a significantly reduced affinity of HSP47-R222S for type I collagen. This altered interaction leads to posttranslational overmodification of type I procollagen produced by dermal fibroblasts, with increased glycosylation and/or hydroxylation of lysine and proline residues as shown by mass spectrometry. Since we also observed a normal intracellular folding and secretion rate of type I procollagen, this overmodification cannot be explained by prolonged exposure of the procollagen molecules to the modifying hydroxyl- and glycosyltransferases, as is commonly observed in other types of OI. We found significant upregulation of several molecular chaperones and enzymes involved in procollagen modification and folding on Western blot and RT-qPCR. In addition, we showed that an imbalance in binding of HSP47-R222S to unfolded type I collagen chains in a gelatin sepharose pulldown assay results in increased binding of other chaperones and modifying enzymes. The elevated expression and binding of this molecular ensemble to type I procollagen suggests a compensatory mechanism for the aberrant binding of HSP47-R222S, eventually leading to overmodification of type I procollagen chains. Together, these results illustrate the importance of HSP47 for proper posttranslational modification and provide insights into the molecular pathomechanisms of the p.(R222S) alteration in HSP47, which leads to a severe OI phenotype., Author summary Heat shock protein 47 (HSP47) is essential for correct collagen folding. We report a homozygous p.(R222S) substitution in HSP47 in a child with severe osteogenesis imperfecta. The highly conserved p.R222 residue is located within the collagen interacting surface and HSP47-R222S shows a significantly reduced affinity for type I collagen. This altered interaction leads to posttranslational overmodification of type I procollagen. In contrast to other types of OI, this overmodification is not caused by prolonged exposure of procollagen to modifying enzymes, since the intracellular folding rate of type I procollagen appears to be normal. We show significant upregulation of several molecular chaperones and collagen-modifying enzymes and increased binding of several of these molecules to unfolded type I collagen chains upon abnormal HSP47-R222S binding. This suggests a compensatory mechanism for aberrant HSP47-R222S binding, eventually leading to overmodification of type I procollagen chains, and underscores the importance of HSP47 for proper posttranslational modification.
- Published
- 2021
26. Matrisome Expression in the Dorsal Root Ganglion
- Author
-
Robin Vroman, Zoë Malfait, Rahel Hunter, Anne-Marie Malfait, Fransiska Malfait, Delfien Syx, and Rachel Miller
- Subjects
Anesthesiology and Pain Medicine ,Neurology ,Neurology (clinical) - Published
- 2022
- Full Text
- View/download PDF
27. Collagens in the Physiopathology of the Ehlers–Danlos Syndromes
- Author
-
Robin Vroman, Fransiska Malfait, Marlies Colman, and Delfien Syx
- Subjects
Peptidylprolyl isomerase ,biology ,Chemistry ,Connective tissue ,Fibrillogenesis ,Tenascin X ,Cell biology ,Extracellular matrix ,ADAMTS2 ,Procollagen peptidase ,medicine.anatomical_structure ,biology.protein ,medicine ,Wound healing - Abstract
The Ehlers–Danlos Syndromes (EDS) comprise a clinically and genetically heterogeneous group of complex hereditary disorders of connective tissue, with common features including joint hypermobility, soft and hyperextensible skin, abnormal wound healing, easy bruising, and signs of generalized connective tissue friability. Initial ultrastructural studies suggested that the abnormalities underlying EDS affected the collagen “wickerwork” of the connective tissue, and early biochemical and genetic studies identified defects in fibrillar types I, III, and V collagen, and in enzymes involved in their posttranslational modification, lysyl hydroxylase 1 and the procollagen amino-proteinase ADAMTS2. More recent discoveries have implicated a range of other, diverse extracellular matrix (ECM) molecules in the physiopathology of EDS, including the glycoprotein tenascin X, the FACIT type XII collagen, the intracellular chaperone and peptidylprolyl isomerase FKBP22, enzymes involved in glycosaminoglycan biosynthesis (D4ST1, DS-epi1, galactosyltransferase I and II), an intracellular zinc transporter ZIP13, (putative) transcription factors ZNF469 and PRDM5, factors involved in the classical complement pathway (C1r and C1s), and most recently, the ECM molecule AEBP1 that is involved in collagen polymerization. In this chapter, we give an overview of the different types of EDS and describe how the identification of their molecular underpinnings, and the study of pathophysiologic consequences of these defects in humans and in cellular and mouse models have provided key insights into the complex pathways of collagen fibrillogenesis and supramolecular organization of the collagen fibrils in the ECM.
- Published
- 2021
- Full Text
- View/download PDF
28. Loss of TANGO1 leads to absence of bone mineralization
- Author
-
Sofie Symoens, Fransiska Malfait, Brecht Guillemyn, Sheela Nampoothiri, and Delfien Syx
- Subjects
COPII VESICLES ,Dentinogenesis imperfecta ,Endocrinology, Diabetes and Metabolism ,Diseases of the musculoskeletal system ,Biology ,COLLAGENOPATHY ,medicine ,Medicine and Health Sciences ,TANGO1 ,Orthopedics and Sports Medicine ,Secretion ,CTAGE5 ,Fibroblast ,Orthopedic surgery ,Endoplasmic reticulum ,Original Articles ,ASSOCIATION ,DEFECTS ,medicine.disease ,Osteochondrodysplasia ,Cell biology ,medicine.anatomical_structure ,COLLAGEN SECRETION ,RC925-935 ,Osteogenesis imperfecta ,OSTEOGENESIS IMPERFECTA ,Original Article ,Type I collagen ,RD701-811 ,Extracellular matrix organization - Abstract
TANGO1 (transport and Golgi organization‐1 homolog) encodes a transmembrane protein, which is located at endoplasmic reticulum (ER) exit sites where it binds bulky cargo, such as collagens, in the lumen and recruits membranes from the ER‐Golgi intermediate compartment (ERGIC) to create an export route for cargo secretion. Mice lacking Mia3 (murine TANGO1 orthologue) show defective secretion of numerous procollagens and lead to neonatal lethality due to insufficient bone mineralization. Recently, aberrant expression of truncated TANGO1 in humans has been shown to cause a mild‐to‐moderate severe collagenopathy associated with dentinogenesis imperfecta, short stature, skeletal abnormalities, diabetes mellitus, and mild intellectual disability. We now show for the first time that complete loss of TANGO1 results in human embryonic lethality with near‐total bone loss and phenocopies the situation of Mia3 −/− mice. Whole‐exome sequencing on genomic DNA (gDNA) of an aborted fetus of Indian descent revealed a homozygous 4‐base pair (4‐bp) deletion in TANGO1 that is heterozygously present in both healthy parents. Parental fibroblast studies showed decreased TANGO1 mRNA expression and protein levels. Type I collagen secretion and extracellular matrix organization were normal, supporting a threshold model for clinical phenotype development. As such, our report broadens the phenotypic and mutational spectrum of TANGO1‐related collagenopathies, and underscores the crucial role of TANGO1 for normal bone development, of which deficiency results in a severe‐to‐lethal form of osteochondrodysplasia. © 2021 American Society for Bone and Mineral Research © 2020 The Authors. JBMR Plus published by Wiley Periodicals LLC. on behalf of American Society for Bone and Mineral Research.
- Published
- 2021
29. b3galt6 Knock-Out Zebrafish Recapitulate β3GalT6-Deficiency Disorders in Human and Reveal a Trisaccharide Proteoglycan Linkage Region
- Author
-
Shuji Mizumoto, Delfien Syx, Charlotte Gistelinck, Andy Willaert, Fransiska Malfait, Fredrik Noborn, Paul Coucke, Göran Larson, Shuhei Yamada, Jan Willem Bek, Adelbert De Clercq, Sarah Delbaere, Lien Alluyn, Phil Salmon, and Bianchet, Mario Antonio
- Subjects
EXPRESSION ,trisaccharide linkage region ,Eherls-Danlos syndrome ,SWIMMING PERFORMANCE ,linkeropathies ,Context (language use) ,PHENOTYPE ,Extracellular matrix ,Glycosaminoglycan ,Collagen fibril organization ,chemistry.chemical_compound ,Cell and Developmental Biology ,CHONDROITIN SULFATE ,Chondroitin sulfate ,Zebrafish ,lcsh:QH301-705.5 ,Original Research ,HEPARAN-SULFATE ,MOLECULAR-CLONING ,BONE MATERIAL PROPERTIES ,IDENTIFICATION ,biology ,GALACTOSYLTRANSFERASE-II ,Biology and Life Sciences ,modeling ,Cell Biology ,Heparan sulfate ,biology.organism_classification ,zebrafish ,COLLAGEN ,Cell biology ,b3galt6 ,chemistry ,Proteoglycan ,lcsh:Biology (General) ,linkage region ,biology.protein ,proteoglycans ,Developmental Biology - Abstract
Proteoglycans are structurally and functionally diverse biomacromolecules found abundantly on cell membranes and in the extracellular matrix. They consist of a core protein linked to glycosaminoglycan chains via a tetrasaccharide linkage region. Here, we show that CRISPR/Cas9-mediatedb3galt6knock-out zebrafish, lacking galactosyltransferase II, which adds the third sugar in the linkage region, largely recapitulate the phenotypic abnormalities seen in human β3GalT6-deficiency disorders. These comprise craniofacial dysmorphism, generalized skeletal dysplasia, skin involvement and indications for muscle hypotonia. In-depth TEM analysis revealed disturbed collagen fibril organization as the most consistent ultrastructural characteristic throughout different affected tissues. Strikingly, despite a strong reduction in glycosaminoglycan content, as demonstrated by anion-exchange HPLC, subsequent LC-MS/MS analysis revealed a small amount of proteoglycans containing a unique linkage region consisting of only three sugars. This implies that formation of glycosaminoglycans with an immature linkage region is possible in a pathogenic context. Our study, therefore unveils a novel rescue mechanism for proteoglycan production in the absence of galactosyltransferase II, hereby opening new avenues for therapeutic intervention.
- Published
- 2020
- Full Text
- View/download PDF
30. Clinical and molecular features of 66 patients with musculocontractural Ehlers-Danlos syndrome caused by pathogenic variants in
- Author
-
Mari, Minatogawa, Ai, Unzaki, Hiroko, Morisaki, Delfien, Syx, Tohru, Sonoda, Andreas R, Janecke, Anne, Slavotinek, Nicol C, Voermans, Yves, Lacassie, Roberto, Mendoza-Londono, Klaas J, Wierenga, Parul, Jayakar, William A, Gahl, Cynthia J, Tifft, Luis E, Figuera, Yvonne, Hilhorst-Hofstee, Alessandra, Maugeri, Ken, Ishikawa, Tomoko, Kobayashi, Yoko, Aoki, Toshihiro, Ohura, Hiroshi, Kawame, Michihiro, Kono, Kosuke, Mochida, Chiho, Tokorodani, Kiyoshi, Kikkawa, Takayuki, Morisaki, Tetsuyuki, Kobayashi, Takaya, Nakane, Akiharu, Kubo, Judith D, Ranells, Ohsuke, Migita, Glenda, Sobey, Anupriya, Kaur, Masumi, Ishikawa, Tomomi, Yamaguchi, Naomichi, Matsumoto, Fransiska, Malfait, Noriko, Miyake, and Tomoki, Kosho
- Subjects
Male ,Phenotype ,Humans ,Abnormalities, Multiple ,Ehlers-Danlos Syndrome ,Female ,Sulfotransferases ,Genetic Association Studies - Abstract
Musculocontractural Ehlers-Danlos syndrome is caused by biallelic loss-of-function variants inWe collected detailed and comprehensive clinical and molecular information regarding previously reported and newly identified patients with mcEDS-Sixty-six patients in 48 families (33 males/females; 0-59 years), including 18 newly reported patients, were evaluated. Japanese was the predominant ethnicity (27 families), associated with three recurrent variants. No apparent genotype-phenotype correlation was noted. Specific craniofacial (large fontanelle with delayed closure, downslanting palpebral fissures and hypertelorism), skeletal (characteristic finger morphologies, joint hypermobility, multiple congenital contractures, progressive talipes deformities and recurrent joint dislocation), cutaneous (hyperextensibility, fine/acrogeria-like/wrinkling palmar creases and bruisability) and ocular (refractive errors) features were observed in most patients (90%). Large subcutaneous haematomas, constipation, cryptorchidism, hypotonia and motor developmental delay were also common (80%). Median ages at the initial episode of dislocation or large subcutaneous haematoma were both 6 years. Nine patients died; their median age was 12 years. Several features, including joint and skin characteristics (hypermobility/extensibility and fragility), were significantly more frequent in patients with mcEDS-This first international collaborative study of mcEDS
- Published
- 2020
31. More than meets the eye: expanding and reviewing the clinical and mutational spectrum of brittle cornea syndrome
- Author
-
Anil Radhakrishnan, Isabelle Migeotte, Fransiska Malfait, Osama Essawi, Serdar Ceylaner, Pelin Ozlem Simsek-Kiper, Sofie Symoens, Tim Van Damme, Laura Muiño Mosquera, Tibbe Dhooge, Brad T. Tinkle, Adila Al Kindy, Maryse Bonduelle, Delfien Syx, Gülen Eda Utine, Sheela Nampoothiri, Pediatrics, Clinical sciences, Vriendenkring VUB, Reproduction and Genetics, and Medical Genetics
- Subjects
Male ,Marfan syndrome ,Génétique clinique ,DNA Mutational Analysis ,Extracellular matrix ,Corneal thinning ,Genotype ,Medicine ,Eye Abnormalities ,PRDM5 ,Child ,Genetics (clinical) ,0303 health sciences ,brittle cornea syndrome ,030305 genetics & heredity ,Pedigree ,DNA-Binding Proteins ,medicine.anatomical_structure ,Osteogenesis imperfecta ,Child, Preschool ,Female ,ZNF469 ,medicine.symptom ,Biologie ,Adult ,Joint Instability ,medicine.medical_specialty ,Adolescent ,Hearing loss ,Connective tissue ,Biology ,03 medical and health sciences ,Young Adult ,Exome Sequencing ,Genetics ,Humans ,Family ,Genetic Association Studies ,030304 developmental biology ,Brittle cornea syndrome ,business.industry ,Infant ,medicine.disease ,Dermatology ,Ehlers–Danlos syndrome ,Mutation ,Skin Abnormalities ,Ehlers-Danlos syndrome ,business ,multisystemic disorder ,Transcription Factors - Abstract
Brittle cornea syndrome (BCS) is a rare autosomal recessive disorder characterized by corneal thinning and fragility, leading to corneal rupture, the main hallmark of this disorder. Non-ocular symptoms include not only hearing loss but also signs of connective tissue fragility, placing it in the Ehlers-Danlos syndrome (EDS) spectrum. It is caused by biallelic pathogenic variants in ZNF469 or PRDM5, which presumably encode transcription factors for extracellular matrix components. We report the clinical and molecular features of nine novel BCS families, four of which harbor variants in ZNF469 and five in PRDM5. We also performed a genotype- and phenotype-oriented literature overview of all (n = 85) reported patients with ZNF469 (n = 53) and PRDM5 (n = 32) variants. Musculoskeletal findings may be the main reason for referral and often raise suspicion of another heritable connective tissue disorder, such as kyphoscoliotic EDS, osteogenesis imperfecta, or Marfan syndrome, especially when a corneal rupture has not yet occurred. Our findings highlight the multisystemic nature of BCS and validate its inclusion in the EDS classification. Importantly, gene panels for heritable connective tissue disorders should include ZNF469 and PRDM5 to allow for timely diagnosis and appropriate preventive measures for this rare condition., SCOPUS: ar.j, info:eu-repo/semantics/published
- Published
- 2020
- Full Text
- View/download PDF
32. Author response for 'Loss of TANGO1 leads to absence of bone mineralization'
- Author
-
Sofie Symoens, Fransiska Malfait, Delfien Syx, Sheela Nampoothiri, and Brecht Guillemyn
- Subjects
medicine.medical_specialty ,Endocrinology ,Chemistry ,Internal medicine ,medicine - Published
- 2020
- Full Text
- View/download PDF
33. b3galt6knock-out zebrafish recapitulate β3GalT6-deficiency disorders in human and reveal a trisaccharide proteoglycan linkage region
- Author
-
Phil Salmon, Sarah Delbaere, Göran Larson, Andy Willaert, Lien Alluyn, Jan Willem Bek, Shuji Mizumoto, Charlotte Gistelinck, Fransiska Malfait, Delfien Syx, Paul Coucke, Adelbert De Clercq, Yamada shuhei, and Fredrik Noborn
- Subjects
Glycosaminoglycan ,Extracellular matrix ,Collagen fibril organization ,Proteoglycan ,biology ,biology.protein ,CRISPR ,Context (language use) ,biology.organism_classification ,Zebrafish ,Phenotype ,Cell biology - Abstract
Proteoglycans are structurally and functionally diverse biomacromolecules found abundantly on cell membranes and in the extracellular matrix. They consist of a core protein linked to glycosaminoglycan chains via a tetrasaccharide linkage region. Here, we show that CRISPR/Cas9-mediatedb3galt6knock-out zebrafish, lacking galactosyltransferase II, which adds the third sugar in the linkage region, largely recapitulate the phenotypic abnormalities seen in human β3GalT6-deficiency disorders. These comprise craniofacial dysmorphism, generalized skeletal dysplasia, skin involvement and indications for muscle hypotonia. In-depth TEM analysis revealed disturbed collagen fibril organization as the most consistent ultrastructural characteristic throughout different affected tissues. Strikingly, despite a strong reduction in glycosaminoglycan content, as demonstrated by anion-exchange HPLC, subsequent LC-MS/MS analysis revealed a small amount of proteoglycans containing a unique linkage region consisting of only three sugars. This implies that formation of glycosaminoglycans with an immature linkage region is possible in a pathogenic context. Our study therefore unveils a novel rescue mechanism for proteoglycan production in the absence of galactosyltransferase II, hereby opening new avenues for therapeutic intervention.
- Published
- 2020
- Full Text
- View/download PDF
34. Bi-allelic AEBP1 mutations in two patients with Ehlers-Danlos syndrome
- Author
-
Nicol C. Voermans, Riet De Rycke, Inge De Wandele, Fransiska Malfait, Olivier Hougrand, Delfien Syx, Sofie Symoens, and Anne De Paepe
- Subjects
Joint hypermobility ,Adult ,Joint Instability ,Male ,Pathology ,medicine.medical_specialty ,Adolescent ,Connective tissue ,Carboxypeptidases ,Biology ,medicine.disease_cause ,Collagen fibril organization ,03 medical and health sciences ,Young Adult ,Genetics ,medicine ,Humans ,Child ,Molecular Biology ,Genetics (clinical) ,Exome sequencing ,Alleles ,Skin ,0303 health sciences ,Mutation ,030305 genetics & heredity ,General Medicine ,Middle Aged ,medicine.disease ,Disorders of movement Donders Center for Medical Neuroscience [Radboudumc 3] ,Phenotype ,Extracellular Matrix ,Repressor Proteins ,medicine.anatomical_structure ,Ehlers–Danlos syndrome ,Child, Preschool ,Skin Abnormalities ,Ehlers-Danlos Syndrome ,Female ,Collagen ,Wound healing - Abstract
Contains fulltext : 207011.pdf (Publisher’s version ) (Closed access) The Ehlers-Danlos syndromes (EDSs) are a clinically and molecularly diverse group of heritable connective tissue disorders caused by defects in a wide range of genes. Recently, bi-allelic loss-of-function mutations in the adipocyte enhancer-binding protein 1 (AEBP1) gene were reported in three families with an autosomal recessive EDS-like condition characterized by thin and hyperextensible skin, poor wound healing with prominent atrophic scarring, joint hypermobility and osteoporosis. Using whole exome sequencing, we identified novel bi-allelic AEBP1 variants in two unrelated adult patients, previously diagnosed with an undefined EDS type, which shows important clinical resemblance to several other EDS subtypes. Our patients present with similar cutaneous and musculoskeletal features as the previously reported patients. They also show unreported clinical features, including pectus deformity, premature aged appearance, sparse and frizzled hair, fatigue and pain. AEBP1 is ubiquitously expressed and encodes the secreted aortic carboxypeptidase-like protein (ACLP) that can bind fibrillar collagens and assist in collagen polymerization. Transmission electron microscopy studies on the patients' skin biopsies show ultrastructural alterations in collagen fibril diameter and appearance, underscoring an important role for ACLP in collagen fibril organization. This report further expands the clinical, molecular and ultrastructural spectrum associated with AEBP1 defects and highlights the complex and variable phenotype associated with this new EDS variant.
- Published
- 2019
- Full Text
- View/download PDF
35. Type III collagen affects dermal and vascular collagen fibrillogenesis and tissue integrity in a mutant Col3a1 transgenic mouse model
- Author
-
Sanne D'hondt, Leen Vanhoutte, Sophie Janssens, Fransiska Malfait, Yoshihiro Ishikawa, Douglas R. Keene, Anne De Paepe, Hans Peter Bächinger, Sofie Symoens, Riet De Rycke, Delfien Syx, Mathieu J.M. Bertrand, Wendy Toussaint, Bart N. Lambrecht, and Brecht Guillemyn
- Subjects
Male ,0301 basic medicine ,Genetically modified mouse ,Heterozygote ,Mutant ,Glycine ,Gene Expression ,Mice, Transgenic ,Fibril ,Tissue Culture Techniques ,Extracellular matrix ,Mice ,03 medical and health sciences ,Sex Factors ,Serine ,medicine ,Animals ,Humans ,Missense mutation ,Molecular Biology ,Skin ,Chemistry ,Fibrillogenesis ,Arteries ,Fibroblasts ,medicine.disease ,Molecular biology ,Disease Models, Animal ,Collagen Type III ,030104 developmental biology ,Amino Acid Substitution ,Ehlers–Danlos syndrome ,Mutation ,Ehlers-Danlos Syndrome ,Female - Abstract
Type III collagen is a major fibrillar collagen consisting of three identical α1(III)-chains that is particularly present in tissues exhibiting elastic properties, such as the skin and the arterial wall. Heterozygous mutations in the COL3A1 gene result in vascular Ehlers-Danlos syndrome (vEDS), a severe, life-threatening disorder, characterized by thin, translucent skin and propensity to arterial, intestinal and uterine rupture. Most human vEDS cases result from a missense mutation substituting a crucial glycine residue in the triple helical domain of the α1(III)-chains. The mechanisms by which these mutant type III collagen molecules cause dermal and vascular fragility are not well understood. We generated a transgenic mouse line expressing mutant type III collagen, containing a typical helical glycine substitution (p.(Gly182Ser)). This Col3a1Tg-G182S mouse line displays a phenotype recapitulating characteristics of human vEDS patients with signs of dermal and vascular fragility. The Col3a1Tg-G182S mice develop severe transdermal skin wounds, resulting in early demise at 13-14weeks of age. We found that this phenotype was associated with a reduced total collagen content and an abnormal collagen III:I ratio, leading to the production of severely malformed collagen fibrils in the extracellular matrix of dermal and arterial tissues. These results indicate that expression of the glycine substitution in the α1(III)-chain disturbs formation of heterotypic type III:I collagen fibrils, and thereby demonstrate a key role for type III collagen in collagen fibrillogenesis in dermal and arterial tissues.
- Published
- 2018
- Full Text
- View/download PDF
36. The Ehlers–Danlos Syndromes against the Backdrop of Inborn Errors of Metabolism
- Author
-
Tim Van Damme, Marlies Colman, Delfien Syx, and Fransiska Malfait
- Subjects
Adult ,Joint Instability ,Adolescent ,inborn errors of metabolism ,Ehlers-Danlos syndromes ,Skin Abnormalities ,Medicine and Health Sciences ,Genetics ,Humans ,Ehlers-Danlos Syndrome ,Connective Tissue Diseases ,Metabolism, Inborn Errors ,pathophysiology ,Genetics (clinical) - Abstract
The Ehlers–Danlos syndromes are a group of multisystemic heritable connective tissue disorders with clinical presentations that range from multiple congenital malformations, over adolescent-onset debilitating or even life-threatening complications of connective tissue fragility, to mild conditions that remain undiagnosed in adulthood. To date, thirteen different EDS types have been recognized, stemming from genetic defects in 20 different genes. While initial biochemical and molecular analyses mainly discovered defects in genes coding for the fibrillar collagens type I, III and V or their modifying enzymes, recent discoveries have linked EDS to defects in non-collagenous matrix glycoproteins, in proteoglycan biosynthesis and in the complement pathway. This genetic heterogeneity explains the important clinical heterogeneity among and within the different EDS types. Generalized joint hypermobility and skin hyperextensibility with cutaneous fragility, atrophic scarring and easy bruising are defining manifestations of EDS; however, other signs and symptoms of connective tissue fragility, such as complications of vascular and internal organ fragility, orocraniofacial abnormalities, neuromuscular involvement and ophthalmological complications are variably present in the different types of EDS. These features may help to differentiate between the different EDS types but also evoke a wide differential diagnosis, including different inborn errors of metabolism. In this narrative review, we will discuss the clinical presentation of EDS within the context of inborn errors of metabolism, give a brief overview of their underlying genetic defects and pathophysiological mechanisms and provide a guide for the diagnostic approach.
- Published
- 2022
- Full Text
- View/download PDF
37. Chronic mucocutaneous candidiasis and connective tissue disorder in humans with impaired JNK1-dependent responses to IL-17A/F and TGF-β
- Author
-
Marco Ritelli, Harry C. Dietz, Danielle T. Avery, Bertrand Boisson, Soraya Boucherit, Lucie Grodecká, Stuart G. Tangye, Romain Lévy, Kathryn Payne, Tomáš Freiberger, Sophie Cypowyj, Juan Li, Valérie Cormier-Daire, Nicoletta Zoppi, Laurent Abel, Geetha Rao, Vivien Béziat, Andrea Guennoun, Benedetta Bigio, Maya Chrabieh, Salim Bougarn, Marina Colombi, Lei Shang, Emilie Corvilain, Yuval Itan, Anne Puel, Franck Rapaport, Nico Marr, Fransiska Malfait, Delfien Syx, Mélanie Migaud, Tanwir Habib, Sabri Boughorbel, Jean-Laurent Casanova, and Cindy S. Ma
- Subjects
Male ,0301 basic medicine ,Connective Tissue Disorder ,MAPK8 ,Immunology ,Mucocutaneous zone ,Connective tissue ,Extracellular matrix ,03 medical and health sciences ,0302 clinical medicine ,Transforming Growth Factor beta ,medicine ,Humans ,Mitogen-Activated Protein Kinase 8 ,Chronic mucocutaneous candidiasis ,Connective Tissue Diseases ,Alleles ,Cells, Cultured ,business.industry ,Candidiasis, Chronic Mucocutaneous ,Interleukin-17 ,General Medicine ,medicine.disease ,030104 developmental biology ,medicine.anatomical_structure ,030220 oncology & carcinogenesis ,Mutation ,Cancer research ,Female ,Signal transduction ,Haploinsufficiency ,business - Abstract
Genetic etiologies of chronic mucocutaneous candidiasis (CMC) disrupt human IL-17A/F-dependent immunity at mucosal surfaces, whereas those of connective tissue disorders (CTDs) often impair the TGF-β-dependent homeostasis of connective tissues. The signaling pathways involved are incompletely understood. We report a three-generation family with an autosomal dominant (AD) combination of CMC and a previously undescribed form of CTD that clinically overlaps with Ehlers-Danlos syndrome (EDS). The patients are heterozygous for a private splice-site variant of MAPK8, the gene encoding c-Jun N-terminal kinase 1 (JNK1), a component of the MAPK signaling pathway. This variant is loss-of-expression and loss-of-function in the patients' fibroblasts, which display AD JNK1 deficiency by haploinsufficiency. These cells have impaired, but not abolished, responses to IL-17A and IL-17F. Moreover, the development of the patients' TH17 cells was impaired ex vivo and in vitro, probably due to the involvement of JNK1 in the TGF-β-responsive pathway and further accounting for the patients' CMC. Consistently, the patients' fibroblasts displayed impaired JNK1- and c-Jun/ATF-2-dependent induction of key extracellular matrix (ECM) components and regulators, but not of EDS-causing gene products, in response to TGF-β. Furthermore, they displayed a transcriptional pattern in response to TGF-β different from that of fibroblasts from patients with Loeys-Dietz syndrome caused by mutations of TGFBR2 or SMAD3, further accounting for the patients' complex and unusual CTD phenotype. This experiment of nature indicates that the integrity of the human JNK1-dependent MAPK signaling pathway is essential for IL-17A- and IL-17F-dependent mucocutaneous immunity to Candida and for the TGF-β-dependent homeostasis of connective tissues.
- Published
- 2019
- Full Text
- View/download PDF
38. Hypomorphic zebrafish models mimic the musculoskeletal phenotype of β4GalT7-deficient Ehlers-Danlos syndrome
- Author
-
Fransiska Malfait, Sofie Symoens, Andy Willaert, Sarah Delbaere, Paul Coucke, Delfien Syx, and Tim Van Damme
- Subjects
0301 basic medicine ,Embryonic Development ,Biology ,Chondrocyte ,03 medical and health sciences ,chemistry.chemical_compound ,Gene Knockout Techniques ,0302 clinical medicine ,Loss of Function Mutation ,medicine ,Animals ,Molecular Biology ,Zebrafish ,Body Patterning ,Gene knockdown ,Cartilage ,Morphant ,Zebrafish Proteins ,biology.organism_classification ,Galactosyltransferases ,Phenotype ,Hypotonia ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Chondroitin sulfate proteoglycan ,030220 oncology & carcinogenesis ,Gene Knockdown Techniques ,Ehlers-Danlos Syndrome ,medicine.symptom - Abstract
β4GalT7 is a transmembrane Golgi enzyme, encoded by B4GALT7, that plays a pivotal role in the proteoglycan linker region formation during proteoglycan biosynthesis. Defects in this enzyme give rise to a rare autosomal recessive form of Ehlers-Danlos syndrome (EDS), currently known as ‘spondylodysplastic EDS (spEDS-B4GALT7)’. This EDS subtype is mainly characterized by short stature, hypotonia and skeletal abnormalities, thereby illustrating its pleiotropic importance during human development. Insights into the pathogenic mechanisms underlying this disabling disease are very limited, in part due to the lack of a relevant in vivo model. As the majority of mutations identified in patients with spEDS-B4GALT7 are hypomorphic, we generated zebrafish models with partial loss of B4galt7 function, including different knockdown (morphant) and mosaic knockout (crispant) b4galt7 zebrafish models and studied the morphologic, functional and molecular aspects in embryonic and larval stages. Morphant and crispant zebrafish show highly similar morphological abnormalities in early development including a small, round head, bowed pectoral fins, short body-axis and mild developmental delay. Several craniofacial cartilage and bone structures are absent or strongly misshapen. In addition, the total amount of sulfated glycosaminoglycans is significantly diminished and particularly heparan and chondroitin sulfate proteoglycan levels are greatly reduced. We also show impaired cartilage patterning and loss of chondrocyte organization in a cartilage-specific Tg(Col2a1aBAC:mcherry) zebrafish reporter line. The occurrence of the same abnormalities in the different models confirms these are specifically caused by B4galt7 deficiency. A disturbed actin pattern, along with a lack of muscle tone, was only noted in morphants in which translation of b4galt7 was blocked. In conclusion, we generated the first viable animal models for spEDS-B4GALT7, and show that in early development the human spEDS-B4GALT7 phenotype is faithfully mimicked in these zebrafish models. Our findings underscore a key role for β4GalT7 in early development of cartilage, bone and muscle. These models will lead to a better understanding of spEDS-B4GALT7 and can be used in future efforts focusing on therapeutic applications.
- Published
- 2019
39. The clinical and mutational spectrum of B3GAT3 linkeropathy: two case reports and literature review
- Author
-
Brecht Guillemyn, Fransiska Malfait, Sofie Symoens, Delfien Syx, Elisabeth Steichen-Gersdorf, Sheela Nampoothiri, Franco Laccone, Marlies Colman, and Tim Van Damme
- Subjects
0301 basic medicine ,Male ,lcsh:Medicine ,HEPARAN-SULFATE PROTEOGLYCANS ,DESBUQUOIS DYSPLASIA ,030105 genetics & heredity ,medicine.disease_cause ,0302 clinical medicine ,B3GAT3 ,Genotype ,Medicine and Health Sciences ,PROTEIN LINKAGE REGION ,Missense mutation ,Pharmacology (medical) ,Joint dislocation ,Glucuronosyltransferase ,Genetics (clinical) ,Glycosaminoglycans ,Genetics ,Mutation ,Homozygote ,XYLT1 MUTATIONS ,General Medicine ,Phenotype ,Connective Tissue ,GAG Linkeropathies ,SKELETAL DYSPLASIA ,Female ,Connective tissue ,BONE FRAGILITY ,medicine.symptom ,Joint hypermobility ,Mutation, Missense ,CORE PROTEIN ,Biology ,Short stature ,03 medical and health sciences ,MISSENSE MUTATION ,medicine ,Humans ,BIOSYNTHESIS ,Genetic Association Studies ,MOLECULAR-CLONING ,Research ,lcsh:R ,Biology and Life Sciences ,medicine.disease ,Dysplasia ,030217 neurology & neurosurgery - Abstract
Background Proteoglycans are large and structurally complex macromolecules which can be found in abundancy in the extracellular matrix and on the surface of all animal cells. Mutations in the genes encoding the enzymes responsible for the formation of the tetrasaccharide linker region between the proteoglycan core protein and the glycosaminoglycan side chains lead to a spectrum of severe and overlapping autosomal recessive connective tissue disorders, collectively coined the ‘glycosaminoglycan linkeropathies’. Results We report the clinical findings of two novel patients with a complex linkeropathy due to biallelic mutations in B3GAT3, the gene that encodes glucuronosyltransferase I, which catalyzes the addition of the ultimate saccharide to the linker region. We identified a previously reported c.667G > A missense mutation and an unreported homozygous c.416C > T missense mutation. We also performed a genotype and phenotype-oriented literature overview of all hitherto reported patients harbouring B3GAT3 mutations. A total of 23 patients from 10 families harbouring bi-allelic mutations and one patient with a heterozygeous splice-site mutation in B3GAT3 have been reported. They all display a complex phenotype characterized by consistent presence of skeletal dysplasia (including short stature, kyphosis, scoliosis and deformity of the long bones), facial dysmorphology, and spatulate distal phalanges. More variably present are cardiac defects, joint hypermobility, joint dislocations/contractures and fractures. Seven different B3GAT3 mutations have been reported, and although the number of patients is still limited, some phenotype-genotype correlations start to emerge. The more severe phenotypes seem to have mutations located in the substrate acceptor subdomain of the catalytic domain of the glucuronosyltransferase I protein while more mildly affected phenotypes seem to have mutations in the NTP-sugar donor substrate binding subdomain. Conclusions Loss-of-function mutations in B3GAT3 are associated with a complex connective tissue phenotype characterized by disproportionate short stature, skeletal dysplasia, facial dysmorphism, spatulate distal phalanges and -to a lesser extent- joint contractures, joint hypermobility with dislocations, cardiac defects and bone fragility. Based on the limited number of reported patients, some genotype-phenotype correlations start to emerge.
- Published
- 2019
40. Novel defects in collagen XII and VI expand the mixed myopathy/Ehlers-Danlos syndrome spectrum and lead to variant-specific alterations in the extracellular matrix
- Author
-
Sofie Symoens, Riet De Rycke, Nathalie Goemans, Delfien Syx, Tibbe Dhooge, Anne Destree, Fransiska Malfait, Florence Petit, Sarah Delbaere, and Olivier Vanakker
- Subjects
Adult ,Collagen Type XII ,Male ,Adolescent ,Decorin ,Collagen Type VI ,Compound heterozygosity ,Tenascin X ,Extracellular matrix ,Muscular Diseases ,Protein Domains ,Collagen VI ,medicine ,Humans ,Child ,Genetics (clinical) ,Cells, Cultured ,biology ,High-Throughput Nucleotide Sequencing ,Tenascin ,Sequence Analysis, DNA ,medicine.disease ,Molecular biology ,Exon skipping ,Extracellular Matrix ,Pedigree ,Fibronectin ,Ehlers–Danlos syndrome ,Child, Preschool ,Mutation ,biology.protein ,Ehlers-Danlos Syndrome ,Female ,Collagen Type V - Abstract
To date, heterozygous or homozygous COL12A1 variants have been reported in 13 patients presenting with a clinical phenotype overlapping with collagen VI–related myopathies and Ehlers–Danlos syndrome (EDS). The small number of reported patients limits thorough investigation of this newly identified syndrome, currently coined as myopathic EDS. DNA from 78 genetically unresolved patients fulfilling the clinical criteria for myopathic EDS was sequenced using a next-generation panel of COL12A1, COL6A1, COL6A2, and COL6A3. Among this cohort, we identified four pathogenic heterozygous in-frame exon skipping (∆) defects in COL12A1, clustering to the thrombospondin N-terminal region and the adjacent collagenous domain (Δ52, Δ53, Δ54, and Δ56 respectively), one heterozygous COL12A1 arginine-to-cysteine substitution of unclear significance (p.(Arg1863Cys)), and compound heterozygous pathogenic COL6A1 variants (c.[98–6G>A];[301C>T]) in one proband. Variant-specific intracellular accumulation of collagen XII chains, extracellular overmodification of the long isoform and near-absence of the short isoform of collagen XII, and extracellular decrease of decorin and tenascin-X were observed for the COL12A1 variants. In contrast, the COL6A1 variants abolished collagen VI and V deposition and increased tenascin-X levels. Our data further support the significant clinical overlap between myopathic EDS and collagen VI–related myopathies, and emphasize the variant-specific consequences of collagen XII defects.
- Published
- 2019
41. RIN2 syndrome: Expanding the clinical phenotype
- Author
-
Ivan Ivanovski, Fransiska Malfait, Simonetta Rosato, Daniela Santodirocco, Marina Beltrami, Livia Garavelli, Delfien Syx, Loredana De Marco, Marzia Pollazzon, and Bert Callewaert
- Subjects
Adult ,Male ,0301 basic medicine ,Connective Tissue Disorder ,Pathology ,medicine.medical_specialty ,Adolescent ,Genotype ,Hearing loss ,Biopsy ,Scoliosis ,Young Adult ,03 medical and health sciences ,Fibrosis ,Genetics ,Guanine Nucleotide Exchange Factors ,Humans ,Medicine ,Abnormalities, Multiple ,Kyphoscoliosis ,Alleles ,Genetic Association Studies ,Genetics (clinical) ,business.industry ,Macrocephaly ,Facies ,Exons ,Syndrome ,Middle Aged ,medicine.disease ,Phenotype ,Radiography ,030104 developmental biology ,Mutation ,Female ,medicine.symptom ,Carrier Proteins ,business ,Cutis laxa - Abstract
Biallelic defects in the RIN2 gene, encoding the Ras and Rab interactor 2 protein, are associated with a rare autosomal recessive connective tissue disorder, with only nine patients from four independent families reported to date. The condition was initially termed MACS syndrome (macrocephaly, alopecia, cutis laxa, and scoliosis), based on the clinical features of the first identified family; however, with the expansion of the clinical phenotype in additional families, it was subsequently coined RIN2 syndrome. Hallmark features of this condition include dysmorphic facial features with striking, progressive facial coarsening, sparse hair, normal to enlarged occipitofrontal circumference, soft redundant and/or hyperextensible skin, and scoliosis. Patients with RIN2 syndrome present phenotypic overlap with other conditions, including EDS (especially the dermatosparaxis and kyphoscoliosis subtypes). Here, we describe a 10th patient, the first patient of Caucasian origin and the oldest reported patient so far, who harbors the previously identified homozygous RIN2 mutation c.1878dupC (p. (Ile627Hisfs*7)). Besides the hallmark features, this patient also presents problems not previously associated with RIN2 syndrome, including cervical vertebral fusion, mild hearing loss, and colonic fibrosis. We provide an overview of the clinical findings in all reported patients with RIN2 mutations and summarize some of the possible pathogenic mechanisms that may underlie this condition. © 2016 Wiley Periodicals, Inc.
- Published
- 2016
- Full Text
- View/download PDF
42. The N‐terminal p.(Ser38Cys) TIMP3 mutation underlying Sorsby fundus dystrophy is a founder mutation disrupting an intramolecular disulfide bond
- Author
-
Julie De Zaeytijd, Delfien Syx, Frauke Coppieters, Frank Peelman, Roosmarijn E. Vandenbroucke, Sarah Naessens, Bart P. Leroy, Frédéric Smeets, and Caroline Van Cauwenbergh
- Subjects
Male ,TIMP3 ,Protein Conformation ,MATRIX METALLOPROTEINASES ,Mutant ,medicine.disease_cause ,PHENOTYPE ,Macular Degeneration ,Protein structure ,Medicine and Health Sciences ,Disulfides ,Genetics (clinical) ,Research Articles ,Genetics ,0303 health sciences ,Mutation ,dimerization ,030305 genetics & heredity ,Phenotype ,Founder Effect ,Pedigree ,Matrix Metalloproteinase 2 ,Female ,Research Article ,EXPRESSION ,glycosylation ,Biology ,03 medical and health sciences ,Structure-Activity Relationship ,TISSUE INHIBITOR ,aberrant disulfide bonding ,medicine ,Humans ,Protein Interaction Domains and Motifs ,030304 developmental biology ,Aged ,Tissue Inhibitor of Metalloproteinase-3 ,STABILITY ,Haplotype ,Wild type ,Biology and Life Sciences ,METALLOPROTEINASES-3 TIMP3 ,Fibroblasts ,GENE ,Molecular Weight ,Gene Expression Regulation ,Haplotypes ,founder mutation ,Sorsby fundus dystrophy ,Founder effect ,Cysteine - Abstract
Sorsby fundus dystrophy (SFD) is a macular degeneration caused by mutations in TIMP3, the majority of which introduce a novel cysteine. However, the exact molecular mechanisms underlying SFD remain unknown. We aimed to provide novel insights into the functional consequences of a distinct N‐terminal mutation. Haplotype reconstruction in three SFD families revealed that the identified c.113C>G, p.(Ser38Cys) mutation is a founder in Belgian and northern French families with a late‐onset SFD phenotype. Functional consequences of the p.(Ser38Cys) mutation were investigated by high‐resolution Western blot analysis of wild type and mutant TIMP3 using patient fibroblasts and in vitro generated proteins, and by molecular modeling of TIMP3 and its interaction partners. We could not confirm a previous hypothesis on dimerization of mutant TIMP3 proteins. However, we identified aberrant intramolecular disulfide bonding. Our data provide evidence for disruption of the established Cys36‐Cys143 disulfide bond and formation of a novel Cys36‐Cys38 bond, possibly associated with increased glycosylation of the protein. In conclusion, we propose a novel pathogenetic mechanism underlying the p.(Ser38Cys) TIMP3 founder mutation involving intramolecular disulfide bonding. These results provide new insights into the pathogenesis of SFD and other retinopathies linked to mutations in TIMP3, such as age‐related macular degeneration.
- Published
- 2019
43. A homozygous pathogenic missense variant broadens the phenotypic and mutational spectrum of CREB3L1-related osteogenesis imperfecta
- Author
-
Sofie Symoens, Brecht Guillemyn, Hülya Kayserili, Fransiska Malfait, Paul Coucke, Delfien Syx, Anne De Paepe, Patrick Sips, and Lynn Demuynck
- Subjects
Male ,Models, Molecular ,PREDICTION ,Mutant ,Vesicular Transport Proteins ,0302 clinical medicine ,Medicine and Health Sciences ,Missense mutation ,Cyclic AMP Response Element-Binding Protein ,COPII ,Genetics (clinical) ,0303 health sciences ,OASIS ,Homozygote ,General Medicine ,Osteogenesis Imperfecta ,Endoplasmic Reticulum Stress ,Pedigree ,Cell biology ,DNA-Binding Proteins ,Phenotype ,Child, Preschool ,Female ,COP-Coated Vesicles ,Protein Binding ,Leucine zipper ,GENES ,Mutation, Missense ,Nerve Tissue Proteins ,Biology ,PROLYL 3-HYDROXYLATION ,Collagen Type I ,MECHANISMS ,03 medical and health sciences ,Protein Domains ,Genetics ,Humans ,Molecular Biology ,Gene ,030304 developmental biology ,COMPLEX ,IDENTIFICATION ,Endoplasmic reticulum ,Biology and Life Sciences ,DNA-binding domain ,SEC23A ,RETICULUM STRESS-RESPONSE ,Astrocytes ,VISUALIZATION ,030217 neurology & neurosurgery ,BRUCK-SYNDROME - Abstract
The cyclic adenosine monophosphate (AMP) responsive element binding protein 3-like 1 (CREB3L1) gene codes for the endoplasmic reticulum stress transducer old astrocyte specifically induced substance (OASIS), which has an important role in osteoblast differentiation during bone development. Deficiency of OASIS is linked to a severe form of autosomal recessive osteogenesis imperfecta (OI), but only few patients have been reported. We identified the first homozygous pathogenic missense variant (p.(Ala304Val)) in a patient with lethal OI, which is located within the highly conserved basic leucine zipper domain, four amino acids upstream of the DNA binding domain. In vitro structural modeling and luciferase assays demonstrate that this missense variant affects a critical residue in this functional domain, thereby decreasing the type I collagen transcriptional binding ability. In addition, overexpression of the mutant OASIS protein leads to decreased transcription of the SEC23A and SEC24D genes, which code for components of the coat protein complex type II (COPII), and aberrant OASIS signaling also results in decreased protein levels of SEC24D. Our findings therefore provide additional proof of the potential involvement of the COPII secretory complex in the context of bone-associated disease.
- Published
- 2019
- Full Text
- View/download PDF
44. Biallelic B3GALT6 mutations cause spondylodysplastic Ehlers–Danlos syndrome
- Author
-
Xiaomeng Pang, Rolph Pfundt, Sofie Symoens, Fransiska Malfait, Marleen Simon, Brecht Guillemyn, Austin Larson, Saskia Bulk, Marion Gérard, Ariana Kariminejad, Sandrine Gulberti, Tim Van Damme, Kathryn C. Chatfield, Olivier Kaye, Delfien Syx, Riet De Rycke, Geneviève Pierquin, Christine E. M. de Die-Smulders, Anne Legrand, Sheela Nampoothiri, Sylvie Fournel-Gigleux, Center for Medical Genetics [Ghent], Ghent University Hospital, Ingénierie Moléculaire et Physiopathologie Articulaire (IMoPA), Université de Lorraine (UL)-Centre National de la Recherche Scientifique (CNRS), Department of Plant Systems Biology, Flanders Institute for Biotechnology, Centre de Rhumatologie, Maastricht University Medical Centre (MUMC), Maastricht University [Maastricht], Department of Human Genetics, Radboud University Medical Center [Nijmegen], Kariminejad-Najmabadi Pathology& Genetics Center, Kariminejad-Najmabadi Pathology & Genetics Center, Amrita Institute of Medical Sciences and Research Center, Hôpital Cochin [AP-HP], Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP), Centre de Génétique Humaine, Université de Liège-CHU Liège, Deaprtment of Clinical Genetics, University Medical Center [Utrecht], Department of Pediatrics [san Diego], UC San Diego School of Medicine, Hôpital Européen Georges Pompidou [APHP] (HEGP), Assistance publique - Hôpitaux de Paris (AP-HP) (AP-HP)-Hôpitaux Universitaires Paris Ouest - Hôpitaux Universitaires Île de France Ouest (HUPO), Service de Génétique [CHU Caen], Université de Caen Normandie (UNICAEN), Normandie Université (NU)-Normandie Université (NU)-CHU Caen, Normandie Université (NU)-Tumorothèque de Caen Basse-Normandie (TCBN)-Tumorothèque de Caen Basse-Normandie (TCBN), Biologie, génétique et thérapies ostéoarticulaires et respiratoires (BIOTARGEN), Normandie Université (NU)-Normandie Université (NU), RS: GROW - R4 - Reproductive and Perinatal Medicine, MUMC+: DA KG Polikliniek (9), and Klinische Genetica
- Subjects
Male ,0301 basic medicine ,MESH: Ehlers-Danlos Syndrome ,IMAGE ,PROTEOGLYCAN ,Gene Expression ,DESBUQUOIS DYSPLASIA ,medicine.disease_cause ,MESH: Galactosyltransferases ,chemistry.chemical_compound ,MESH: Child ,Child ,MESH: High-Throughput Nucleotide Sequencing ,Genetics (clinical) ,Exome sequencing ,Mutation ,biology ,XYLT1 MUTATIONS ,High-Throughput Nucleotide Sequencing ,General Medicine ,Heparan sulfate ,Galactosyltransferases ,Phenotype ,MESH: Infant ,3. Good health ,Child, Preschool ,Female ,SKELETAL DYSPLASIA ,BONE FRAGILITY ,Adult ,MESH: Mutation ,MESH: Gene Expression ,[SDV.CAN]Life Sciences [q-bio]/Cancer ,MESH: Phenotype ,PATIENT ,Collagen fibril organization ,03 medical and health sciences ,All institutes and research themes of the Radboud University Medical Center ,MESH: Whole Exome Sequencing ,MESH: Enzyme Assays ,Exome Sequencing ,Genetics ,medicine ,Humans ,Molecular Biology ,Enzyme Assays ,HEPARAN-SULFATE ,Spondyloepimetaphyseal dysplasia ,SPECTRUM ,[SDV.GEN]Life Sciences [q-bio]/Genetics ,Neurodevelopmental disorders Donders Center for Medical Neuroscience [Radboudumc 7] ,MESH: Humans ,MESH: Child, Preschool ,Infant ,MESH: Adult ,medicine.disease ,Molecular biology ,B3GAT3 MUTATION ,MESH: Male ,030104 developmental biology ,Proteoglycan ,chemistry ,[SDV.GEN.GH]Life Sciences [q-bio]/Genetics/Human genetics ,Ehlers–Danlos syndrome ,biology.protein ,Ehlers-Danlos Syndrome ,MESH: Female - Abstract
International audience; Proteoglycans are among the most abundant and structurally complex biomacromolecules and play critical roles in connective tissues. They are composed of a core protein onto which glycosaminoglycan (GAG) side chains are attached via a linker region. Biallelic mutations in B3GALT6, encoding one of the linker region glycosyltransferases, are known to cause either spondyloepimetaphyseal dysplasia (SEMD) or a severe pleiotropic form of Ehlers-Danlos syndromes (EDS). This study provides clinical, molecular and biochemical data on 12 patients with biallelic B3GALT6 mutations. Notably, all patients have features of both EDS and SEMD. In addition, some patients have severe and potential life-threatening complications such as aortic dilatation and aneurysm, cervical spine instability and respiratory insufficiency. Whole-exome sequencing, next generation panel sequencing and direct sequencing identified biallelic B3GALT6 mutations in all patients. We show that these mutations reduce the amount of β3GalT6 protein and lead to a complete loss of galactosyltransferase activity. In turn, this leads to deficient GAG synthesis, and ultrastructural abnormalities in collagen fibril organization. In conclusion, this study redefines the phenotype associated with B3GALT6 mutations on the basis of clinical, molecular and biochemical data in 12 patients, and provides an in-depth assessment of β3GalT6 activity and GAG synthesis to better understand this rare condition.
- Published
- 2018
- Full Text
- View/download PDF
45. Peripheral Mechanisms Contributing to Osteoarthritis Pain
- Author
-
P.B. Tran, Delfien Syx, Rachel E. Miller, and Anne-Marie Malfait
- Subjects
Nociception ,medicine.medical_specialty ,Arthritis ,Inflammation ,Osteoarthritis ,Bioinformatics ,Article ,03 medical and health sciences ,0302 clinical medicine ,Rheumatology ,Internal medicine ,Neuroplasticity ,medicine ,Animals ,Humans ,Neurons, Afferent ,Sensitization ,030203 arthritis & rheumatology ,Analgesics ,business.industry ,medicine.disease ,Arthritis, Experimental ,Immunity, Innate ,medicine.anatomical_structure ,Nociceptor ,Cytokines ,medicine.symptom ,Chronic Pain ,business ,030217 neurology & neurosurgery - Abstract
PURPOSE OF REVIEW: Osteoarthritis (OA) is the most common form of arthritis and a major source of pain and disability worldwide. OA-associated pain is usually refractory to classically used analgesics, and disease-modifying therapies are still lacking. Therefore, a better understanding of mechanisms and mediators contributing to the generation and maintenance of OA pain is critical for the development of efficient and safe pain-relieving therapies. RECENT FINDINGS: Both peripheral and central mechanisms contribute to OA pain. Clinical evidence suggests that a strong peripheral nociceptive drive from the affected joint maintains pain and central sensitization associated with OA. Mediators present in the OA joint, including nerve growth factor, chemokines, cytokines, and inflammatory cells can contribute to sensitization. Furthermore, structural alterations in joint innervation and nerve damage occur in the course of OA. SUMMARY: Several interrelated pathological processes, including joint damage, structural reorganization of joint afferents, low-grade inflammation, neuroplasticity and nerve damage all contribute to the pain observed in OA. It can be anticipated that elucidating exactly how these mechanisms are operational in the course of progressive OA may lead to the identification of novel targets for intervention.
- Published
- 2018
46. Genetic Defects in TAPT1 Disrupt Ciliogenesis and Cause a Complex Lethal Osteochondrodysplasia
- Author
-
Martine Biervliet, Elena Makareeva, Aileen M. Barnes, Anne De Paepe, Andy Willaert, Fransiska Malfait, Wouter Steyaert, Joan C. Marini, Sanne D'hondt, Eckhard Witten, Sofie Symoens, Kris Vleminckx, Paul Coucke, Julie De Backer, Brecht Guillemyn, Ann Huysseune, Gabriele Gillessen-Kaesbach, Delfien Syx, Sergey Leikin, Charlotte Gistelinck, Surgical clinical sciences, and Clinical sciences
- Subjects
Male ,Embryo, Nonmammalian ,Molecular Sequence Data ,Ciliary basal body ,Signal transduction ,Osteochondrodysplasias ,Ciliopathies ,Article ,Craniofacial Abnormalities ,Neural crest ,Cranial neural crest ,Cell Movement ,Ciliogenesis ,Genetics ,medicine ,Animals ,Humans ,Genetics(clinical) ,Amino Acid Sequence ,Cilia ,Zebrafish ,Genetics (clinical) ,Body Patterning ,Sequence Homology, Amino Acid ,biology ,Ossification, Heterotopic ,Research Support, Non-U.S. Gov't ,Cilium ,Gene Expression Regulation, Developmental ,Membrane Proteins ,Cell Differentiation ,zebrafish ,Research Support, N.I.H., Intramural ,biology.organism_classification ,medicine.disease ,Osteochondrodysplasia ,Pedigree ,Cell biology ,protein transport ,Female ,mutation ,In situ hybridization ,Ciliary Motility Disorders - Abstract
The evolutionarily conserved transmembrane anterior posterior transformation 1 protein, encoded by TAPT1, is involved in murine axial skeletal patterning, but its cellular function remains unknown. Our study demonstrates that TAPT1 mutations underlie a complex congenital syndrome, showing clinical overlap between lethal skeletal dysplasias and ciliopathies. This syndrome is characterized by fetal lethality, severe hypomineralization of the entire skeleton and intra-uterine fractures, and multiple congenital developmental anomalies affecting the brain, lungs, and kidneys. We establish that wild-type TAPT1 localizes to the centrosome and/or ciliary basal body, whereas defective TAPT1 mislocalizes to the cytoplasm and disrupts Golgi morphology and trafficking and normal primary cilium formation. Knockdown of tapt1b in zebrafish induces severe craniofacial cartilage malformations and delayed ossification, which is shown to be associated with aberrant differentiation of cranial neural crest cells.
- Published
- 2015
- Full Text
- View/download PDF
47. Defective Proteolytic Processing of Fibrillar Procollagens and Prodecorin Due to BiallelicBMP1Mutations Results in a Severe, Progressive Form of Osteogenesis Imperfecta
- Author
-
Sofie Symoens, Delfien Syx, Margo L. Whiteford, Ana Berta Sousa, Ana Medeira, Anne De Paepe, Fransiska Malfait, Paul Coucke, Brecht Guillemyn, and Trinh Hermanns-Lê
- Subjects
Genetics ,Endocrinology, Diabetes and Metabolism ,Fibrillogenesis ,Biology ,medicine.disease ,Compound heterozygosity ,Molecular biology ,Bone morphogenetic protein 1 ,Frameshift mutation ,Extracellular matrix ,Procollagen peptidase ,Osteogenesis imperfecta ,medicine ,Missense mutation ,Orthopedics and Sports Medicine - Abstract
Whereas the vast majority of osteogenesis imperfecta (OI) is caused by autosomal dominant defects in the genes encoding type I procollagen, mutations in a myriad of genes affecting type I procollagen biosynthesis or bone formation and homeostasis have now been associated with rare autosomal recessive OI forms. Recently, homozygous or compound heterozygous mutations in BMP1, encoding the metalloproteases bone morphogenetic protein-1 (BMP1) and its longer isoform mammalian Tolloid (mTLD), were identified in 5 children with a severe autosomal recessive form of OI and in 4 individuals with mild to moderate bone fragility. BMP1/mTLD functions as the procollagen carboxy-(C)-proteinase for types I to III procollagen but was also suggested to participate in amino-(N)-propeptide cleavage of types V and XI procollagens and in proteolytic trimming of other extracellular matrix (ECM) substrates. We report the phenotypic characteristics and natural history of 4 adults with severe, progressive OI characterized by numerous fractures, short stature with rhizomelic shortening, and deformity of the limbs and variable kyphoscoliosis, in whom we identified novel biallelic missense and frameshift mutations in BMP1. We show that BMP1/mTLD-deficiency in humans not only results in delayed cleavage of the type I procollagen C-propeptide but also hampers the processing of the small leucine-rich proteoglycan prodecorin, a regulator of collagen fibrillogenesis. Immunofluorescent staining of types I and V collagen and transmission electron microscopy of the dermis show impaired assembly of heterotypic type I/V collagen fibrils in the ECM. Our study thus highlights the severe and progressive nature of BMP1-associated OI in adults and broadens insights into the functional consequences of BMP1/mTLD-deficiency on ECM organization.
- Published
- 2015
- Full Text
- View/download PDF
48. Genetics of the Ehlers–Danlos syndrome: more than collagen disorders
- Author
-
Sofie Symoens, Anne De Paepe, Fransiska Malfait, Paul Coucke, Delfien Syx, and Tim Van Damme
- Subjects
Joint hypermobility ,Genetics ,Pathology ,medicine.medical_specialty ,biology ,Health Policy ,Connective tissue ,medicine.disease ,Tenascin X ,Pathogenesis ,Extracellular matrix ,medicine.anatomical_structure ,Osteogenesis imperfecta ,Ehlers–Danlos syndrome ,Collagen disorder ,medicine ,biology.protein ,Pharmacology (medical) ,Pharmacology, Toxicology and Pharmaceutics (miscellaneous) - Abstract
Introduction: The Ehlers-Danlos syndrome (EDS) comprises a clinically and genetically diverse group of heritable connective tissue disorders, characterized by joint hypermobility, skin hyperextensibility and generalized connective tissue friability. Although the initially characterized subtypes were shown to result from defects in fibrillar collagens (types I, III, V) or their modifying enzymes, recent discoveries have implicated other molecules, such as tenascin X and glycosaminoglycan synthesis enzymes, in the pathogenesis of these disorders.Areas covered: This article summarizes the current knowledge on the biosynthesis of collagen fibrils and focuses on the molecules involved in this process, especially those relevant to the pathogenesis of EDS. It also provides an overview of the general clinical presentation of EDS and the genetic defects underlying its different subtypes.Expert opinion: The recent identification of several novel types of EDS has greatly expanded its clinical and genetic heterogenei...
- Published
- 2015
- Full Text
- View/download PDF
49. Hypermobility, the Ehlers-Danlos syndromes and chronic pain
- Author
-
Delfien, Syx, Inge, De Wandele, Lies, Rombaut, and Fransiska, Malfait
- Subjects
Joint Instability ,Humans ,Ehlers-Danlos Syndrome ,Chronic Pain - Abstract
Chronic widespread pain is a common complaint among individuals affected by generalised joint hypermobility. In the absence of other conditions that cause chronic pain, these individuals are usually diagnosed with joint hypermobility syndrome (JHS). JHS is a multifactorial trait with a strong genetic basis, but no specific genetic markers. Clinical overlap of JHS is seen with heritable connective tissue disorders, particularly with the Ehlers-Danlos syndrome, hypermobile type (hEDS). The Ehlers-Danlos syndromes (EDS) comprise a heterogeneous group of rare monogenic conditions that are characterised by joint hypermobility, skin and vascular fragility and generalised connective tissue friability, and are caused by genetic defects in an array of extracellular matrix genes. The genetic basis of hEDS remains however unknown, in contrast to other well-described EDS subtypes. In view of the considerable clinical overlap with JHS, many consider it and hEDS to be a single clinical entity. Clinical experience and a limited number of clinical studies show that chronic pain also is common in EDS patients, especially in hEDS. The specific underlying causes and mechanisms of pain in JHS and EDS remain poorly understood. Factors likely contributing to the generation and chronicity of pain include nociceptive pain, directly based on structural changes in affected joints, muscle and connective tissue; neuropathic pain; impaired proprioception and muscle weakness; and central sensitisation. These mechanisms are not mutually exclusive, and likely more than one mechanism may be present. Furthermore, anxiety, depression, and other variables may influence the phenotype. Chronic pain in JHS and EDS patients often is inadequately controlled by traditional analgesics and physical therapy. In view of the high prevalence of these underrecognised conditions, future studies addressing the nature and mediators of chronic pain are needed in order to potentially identify novel targets for therapeutic intervention and optimise treatment.
- Published
- 2017
50. Pain-related behaviors and abnormal cutaneous innervation in a murine model of classical Ehlers-Danlos syndrome.
- Author
-
Delfien Syx, Miller, Rachel E., Obeidat, Alia M., Tran, Phuong B., Vroman, Robin, Malfait, Zöe, Mille, Richard J., Malfait, Fransiska, Malfait, Anne-Marie, Syx, Delfien, Malfait, Zoë, and Miller, Richard J
- Published
- 2020
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.