Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 50
Filter
1.
J Med Genet ; 61(3): 232-238, 2024 Feb 21.
Article in English | MEDLINE | ID: mdl-37813462

ABSTRACT

BACKGROUND: The Ehlers-Danlos syndromes (EDS) are heritable disorders of connective tissue (HDCT), reclassified in the 2017 nosology into 13 subtypes. The genetic basis for hypermobile Ehlers-Danlos syndrome (hEDS) remains unknown. METHODS: Whole exome sequencing (WES) was undertaken on 174 EDS patients recruited from a national diagnostic service for complex EDS and a specialist clinic for hEDS. Patients had already undergone expert phenotyping, laboratory investigation and gene sequencing, but were without a genetic diagnosis. Filtered WES data were reviewed for genes underlying Mendelian disorders and loci reported in EDS linkage, transcriptome and genome-wide association studies (GWAS). A genetic burden analysis (Minor Allele Frequency (MAF) <0.05) incorporating 248 Avon Longitudinal Study of Parents and Children (ALSPAC) controls sequenced as part of the UK10K study was undertaken using TASER methodology. RESULTS: Heterozygous pathogenic (P) or likely pathogenic (LP) variants were identified in known EDS and Loeys-Dietz (LDS) genes. Multiple variants of uncertain significance where segregation and functional analysis may enable reclassification were found in genes associated with EDS, LDS, heritable thoracic aortic disease (HTAD), Mendelian disorders with EDS symptomatology and syndromes with EDS-like features. Genetic burden analysis revealed a number of novel loci, although none reached the threshold for genome-wide significance. Variants with biological plausibility were found in genes and pathways not currently associated with EDS or HTAD. CONCLUSIONS: We demonstrate the clinical utility of large panel-based sequencing and WES for patients with complex EDS in distinguishing rare EDS subtypes, LDS and related syndromes. Although many of the P and LP variants reported in this cohort would be identified with current panel testing, they were not at the time of this study, highlighting the use of extended panels and WES as a clinical tool for complex EDS. Our results are consistent with the complex genetic architecture of EDS and suggest a number of novel hEDS and HTAD candidate genes and pathways.


Subject(s)
Connective Tissue Diseases , Ehlers-Danlos Syndrome , Child , Humans , Genome-Wide Association Study , Longitudinal Studies , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics
2.
Am J Hum Genet ; 108(7): 1342-1349, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34143952

ABSTRACT

EDEM3 encodes a protein that converts Man8GlcNAc2 isomer B to Man7-5GlcNAc2. It is involved in the endoplasmic reticulum-associated degradation pathway, responsible for the recognition of misfolded proteins that will be targeted and translocated to the cytosol and degraded by the proteasome. In this study, through a combination of exome sequencing and gene matching, we have identified seven independent families with 11 individuals with bi-allelic protein-truncating variants and one individual with a compound heterozygous missense variant in EDEM3. The affected individuals present with an inherited congenital disorder of glycosylation (CDG) consisting of neurodevelopmental delay and variable facial dysmorphisms. Experiments in human fibroblast cell lines, human plasma, and mouse plasma and brain tissue demonstrated decreased trimming of Man8GlcNAc2 isomer B to Man7GlcNAc2, consistent with loss of EDEM3 enzymatic activity. In human cells, Man5GlcNAc2 to Man4GlcNAc2 conversion is also diminished with an increase of Glc1Man5GlcNAc2. Furthermore, analysis of the unfolded protein response showed a reduced increase in EIF2AK3 (PERK) expression upon stimulation with tunicamycin as compared to controls, suggesting an impaired unfolded protein response. The aberrant plasma N-glycan profile provides a quick, clinically available test for validating variants of uncertain significance that may be identified by molecular genetic testing. We propose to call this deficiency EDEM3-CDG.


Subject(s)
Calcium-Binding Proteins/genetics , Congenital Disorders of Glycosylation/genetics , Endoplasmic Reticulum/genetics , alpha-Mannosidase/genetics , Adolescent , Alleles , Calcium-Binding Proteins/deficiency , Cell Line , Child , Child, Preschool , Congenital Disorders of Glycosylation/blood , Developmental Disabilities/genetics , Female , Glycoproteins/blood , Glycosylation , Humans , Infant , Intellectual Disability/genetics , Male , Mutation , Pedigree , Polysaccharides/blood , Proteostasis Deficiencies/genetics , alpha-Mannosidase/deficiency
3.
Pract Neurol ; 24(2): 90-97, 2024 Mar 19.
Article in English | MEDLINE | ID: mdl-38160052

ABSTRACT

Ehlers-Danlos syndromes (EDS) is an umbrella term describing 14 types, of which 13 are rare and monogenic, with overlapping features of joint hypermobility, skin, and vascular fragility, and generalised connective tissue friability. Hypermobile EDS currently has no identified genetic cause. Most of the rare monogenic EDS types can have neurological features, which are often part of major or minor diagnostic criteria for each type. This review aims to highlight the neurological features and other key characteristics of these EDS types. This should improve recognition of these features, enabling more timely consideration and confirmation or exclusion through genetic testing. In practice, many healthcare professionals still refer to patients as having 'EDS'. However, the different EDS types have distinct clinical features as well as different underlying genetic causes and pathogenic mechanisms, and each requires bespoke management and surveillance. Defining the EDS type is therefore crucial, as EDS is not in itself a diagnosis.


Subject(s)
Ehlers-Danlos Syndrome , Joint Instability , Vascular Diseases , Humans , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/pathology , Genetic Testing , Joint Instability/diagnosis , Joint Instability/genetics
4.
Am J Hum Genet ; 107(5): 989-999, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33053334

ABSTRACT

Osteogenesis imperfecta (OI) is characterized primarily by susceptibility to fractures with or without bone deformation. OI is genetically heterogeneous: over 20 genetic causes are recognized. We identified bi-allelic pathogenic KDELR2 variants as a cause of OI in four families. KDELR2 encodes KDEL endoplasmic reticulum protein retention receptor 2, which recycles ER-resident proteins with a KDEL-like peptide from the cis-Golgi to the ER through COPI retrograde transport. Analysis of patient primary fibroblasts showed intracellular decrease of HSP47 and FKBP65 along with reduced procollagen type I in culture media. Electron microscopy identified an abnormal quality of secreted collagen fibrils with increased amount of HSP47 bound to monomeric and multimeric collagen molecules. Mapping the identified KDELR2 variants onto the crystal structure of G. gallus KDELR2 indicated that these lead to an inactive receptor resulting in impaired KDELR2-mediated Golgi-ER transport. Therefore, in KDELR2-deficient individuals, OI most likely occurs because of the inability of HSP47 to bind KDELR2 and dissociate from collagen type I. Instead, HSP47 remains bound to collagen molecules extracellularly, disrupting fiber formation. This highlights the importance of intracellular recycling of ER-resident molecular chaperones for collagen type I and bone metabolism and a crucial role of HSP47 in the KDELR2-associated pathogenic mechanism leading to OI.


Subject(s)
Bone and Bones/metabolism , Collagen Type I/metabolism , HSP47 Heat-Shock Proteins/metabolism , Osteogenesis Imperfecta/genetics , Vesicular Transport Proteins/metabolism , Adult , Alleles , Amino Acid Sequence , Animals , Binding Sites , Bone and Bones/pathology , Chickens , Child, Preschool , Collagen Type I/chemistry , Collagen Type I/genetics , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Golgi Apparatus/metabolism , Golgi Apparatus/pathology , HSP47 Heat-Shock Proteins/chemistry , HSP47 Heat-Shock Proteins/genetics , Humans , Infant , Male , Osteogenesis Imperfecta/diagnosis , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Pedigree , Primary Cell Culture , Protein Binding , Protein Interaction Domains and Motifs , Protein Structure, Secondary , Protein Transport , Sequence Alignment , Sequence Homology, Amino Acid , Vesicular Transport Proteins/chemistry , Vesicular Transport Proteins/genetics
5.
Clin Exp Rheumatol ; 40 Suppl 134(5): 46-62, 2022 May.
Article in English | MEDLINE | ID: mdl-35587586

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.


Subject(s)
Collagen Type I, alpha 1 Chain , Ehlers-Danlos Syndrome , Collagen , Collagen Type III/genetics , Ehlers-Danlos Syndrome/complications , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics , Humans , Mutation , Phenotype
6.
J Clin Periodontol ; 49(12): 1244-1252, 2022 12.
Article in English | MEDLINE | ID: mdl-35833531

ABSTRACT

AIM: Periodontal Ehlers-Danlos syndrome (pEDS) is a monogenic type of Ehlers-Danlos syndrome characterized by periodontal destruction at a young age. The present study aimed to document the oral phenotype of pEDS based on prospective clinical investigations. MATERIALS AND METHODS: Thirty-five adult individuals from 13 families with a clinically and genetically confirmed diagnosis of pEDS underwent a systematic oral assessment. RESULTS: Periodontitis stage 3 or 4 or edentulism due to periodontal destruction were diagnosed in 94% of the individuals. First permanent tooth loss was reported at the age of 21.5 years (median; range 13-43 years). Deep periodontal pockets were infrequent, with 94% measuring <4 mm. However, there was increased clinical attachment loss (CAL) averaging 8 mm (range 4-13 mm), and the probability of being edentate between the age of 35 and 44 years was 28-47% compared with less than 0.25% of the general population. Radiographic anomalous findings were only found in a portion of subjects and consisted of fused roots of maxillary second molars (81%), root hypoplasia (57%), taurodontism (26%) and tooth rotation of premolars (67%). As such, radiographic findings are not considered common characteristics of pEDS. CONCLUSIONS: Characteristic oral traits of pEDS in adults are severe CAL with shallow probing depths and marked gingival recession. This is complemented by a lack of attached gingiva. These indications need to be paralleled by genetic analyses to diagnose pEDS unambiguously.


Subject(s)
Ehlers-Danlos Syndrome , Gingival Recession , Periodontitis , Humans , Ehlers-Danlos Syndrome/complications , Prospective Studies , Gingival Recession/etiology , Bicuspid , Periodontal Attachment Loss
7.
Genet Med ; 23(2): 316-322, 2021 02.
Article in English | MEDLINE | ID: mdl-33005042

ABSTRACT

PURPOSE: We report prospective clinical investigations of children affected with periodontal Ehlers-Danlos syndrome (pEDS). The main clinical features of pEDS in adults are early severe periodontitis, generalized lack of attached gingiva, and pretibial hemosiderin plaques due to dominant pathogenic variants in the C1R or C1S genes. METHODS: Nineteen children with a parent diagnosed with molecularly confirmed pEDS underwent physical examination including oral and radiological investigations followed by genetic testing. RESULTS: The only consistent manifestation of pEDS in childhood was a characteristic gingival phenotype: generalized lack of attached gingiva. All children with this gingival phenotype had inherited the familial pathogenic variant (n = 12) whereas the gingival phenotype was absent in children without the familial pathogenic variant (n = 7). Easy bruising was reported in eight affected and zero unaffected children. Other manifestations of pEDS were rarely present in children. Only 2/12 affected children aged 8 and 13 years fulfilled the clinical criteria for pEDS. CONCLUSION: Generalized lack of attached gingiva is a pathognomonic feature of pEDS and the only clinical finding that is consistently present in affected adults and children. This is important because an early diagnosis may facilitate better dental hygiene in childhood, which may be essential to prevent early dental loss.


Subject(s)
Ehlers-Danlos Syndrome , Periodontitis , Adult , Child , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics , Gingiva , Humans , Phenotype , Prospective Studies
8.
J Med Genet ; 57(11): 769-776, 2020 11.
Article in English | MEDLINE | ID: mdl-32467296

ABSTRACT

BACKGROUND: The Ehlers-Danlos syndromes (EDS) are a group of connective tissue disorders with several recognised types. Patients with a type of EDS have connective tissue abnormalities resulting in a varying degree of joint hypermobility, skin and vascular fragility and generalised tissue friability. Classical EDS (cEDS) typically occurs as a result of dominant pathogenic variants in COL5A1 or COL5A2. The cardinal features of cEDS are hyperextensible skin, atrophic scarring and joint hypermobility. Arterial complications are more characteristically a feature of vascular EDS although individual cases of arterial events in cEDS have been reported. METHODS: A cohort of 154 patients with a clinical diagnosis of cEDS from the UK was analysed. RESULTS: Seven patients (4.5%) with a diagnosis of cEDS (four pathogenic, one likely pathogenic and two variants of uncertain significance in COL5A1) who had experienced arterial complications were identified. Arterial complications mostly involved medium-sized vessels and also two abdominal aortic aneurysms. No unique clinical features were identified in this group of patients. CONCLUSION: There is a possible increased risk of arterial complications in patients with cEDS, although not well-defined. Clinicians need to be aware of this possibility when presented with a patient with an arterial complication and features of cEDS. Long-term management in families with cEDS and a vascular complication should be individually tailored to the patient's history and their family's history of vascular events.


Subject(s)
Collagen Type V/genetics , Connective Tissue Diseases/genetics , Ehlers-Danlos Syndrome/genetics , Adult , Aged , Connective Tissue Diseases/pathology , Female , Genetic Predisposition to Disease , Humans , Joint Instability/genetics , Joint Instability/pathology , Male , Middle Aged , Mutation/genetics , Skin Abnormalities/genetics , Skin Abnormalities/pathology
9.
Genet Med ; 22(10): 1576-1582, 2020 10.
Article in English | MEDLINE | ID: mdl-32572181

ABSTRACT

PURPOSE: Currently, 31 patients with classical-like EDS (clEDS) due to tenascin-X deficiency have been reported in the literature. We report on the clinical and molecular characteristics of 20 additional patients with clEDS to expand knowledge and to enable improved management of this rare genetic disorder. METHODS: Patients diagnosed with clEDS by the national EDS service in the UK (n = 21) and abroad (n = 1) were asked for consent for publication of their clinical and molecular data. RESULTS: Of 22 patients, 20 consented. All patients had typical features of clEDS: joint hypermobility, easy bruising, and skin hyperextensibility without atrophic scars. Importantly, 3/20 patients experienced gastrointestinal complications consisting of small or large bowel ruptures and one esophageal rupture. Other notable observations included two separate occurrences of spontaneous compartment syndrome, suspicion of nonaccidental injury due to significant bruising, and significant clinical variability regarding the debilitating effect of joint dislocations. CONCLUSIONS: We propose a predisposition to tissue fragility, particularly of the gastrointestinal tract in patients with clEDS. As such, clinical and molecular confirmation of this diagnosis is essential. It is recommended to follow up these patients closely to understand the natural history to develop better recommendations for management.


Subject(s)
Ehlers-Danlos Syndrome , Joint Instability , Skin Abnormalities , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics , Extracellular Matrix , Humans , Joint Instability/diagnosis , Joint Instability/genetics
10.
Am J Med Genet A ; 182(3): 553-556, 2020 03.
Article in English | MEDLINE | ID: mdl-31833208

ABSTRACT

Familial amniotic band sequence (ABS) is rare but has been reported in the offspring of mothers with connective tissue disorders. We present a family of two half-siblings with ABS who share the same biological father. Following a serious vascular event a de novo pathogenic variant in COL3A1 was detected in the father, confirming a diagnosis of vascular Ehlers-Danlos syndrome (vEDS). The same variant was found in both his ABS-affected children but not in his unaffected child. The amniotic membrane is derived from fetal tissue, type III collagen being a component. As the affected children are paternal half-siblings, ABS was less likely due to maternal factors. Rather, the amniotic bands may have resulted from decreased type III collagen production as seen in people with vEDS, causing fragility of the amniotic membrane. Consequently, it is important to consider vEDS in patients with ABS.


Subject(s)
Amniotic Band Syndrome/genetics , Collagen Type III/genetics , Ehlers-Danlos Syndrome/genetics , Adult , Amniotic Band Syndrome/complications , Amniotic Band Syndrome/diagnosis , Amniotic Band Syndrome/pathology , Child , Connective Tissue Diseases/complications , Connective Tissue Diseases/diagnosis , Connective Tissue Diseases/genetics , Connective Tissue Diseases/pathology , Ehlers-Danlos Syndrome/complications , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/pathology , Female , Humans , Infant, Newborn , Male , Mutation/genetics , Paternal Inheritance/genetics , Phenotype , Siblings
11.
Am J Med Genet A ; 182(5): 994-1007, 2020 05.
Article in English | MEDLINE | ID: mdl-32091183

ABSTRACT

Arthrochalasia Ehlers-Danlos syndrome (aEDS) is a rare autosomal dominant connective tissue disorder that is characterized by congenital bilateral hip dislocations, severe generalized joint hypermobility, recurrent joint (sub)luxations, and skin hyperextensibility. To date, 42 patients with aEDS have been published. We report 12 patients with aEDS from 10 families with 6 unpublished individuals and follow-up data on 6 adult patients. The clinical features are largely comparable with patients reported in the literature. Most (n = 10) patients had variants leading to (partial) loss of exon 6 of the COL1A1 or COL1A2 genes. One patient did not have a previously reported likely pathogenic COL1A1 variant. Data regarding management were retrieved. Hip surgery was performed in 5/12 patients and 3/12 patients underwent spinal surgery. As much as 4/12 patients were wheelchair-bound or unable to walk unaided. Fractures were present in 9/12 individuals with 1 patient requiring bisphosphonate treatment. Echocardiograms were performed in 10 patients and 2 individuals showed an abnormality likely unrelated to aEDS. One patient gave birth to two affected children and went through preterm labor requiring medication but had no additional complications. Of the eight adults in our cohort, the majority entered a career. Our data point toward a genotype-phenotype relationship with individuals with aEDS due to pathogenic COL1A1 variants causing complete or partial loss of exon 6 being more severely affected regarding musculoskeletal features. There is a significant lack of knowledge with regard to management of aEDS, particularly in adulthood. As such, systematic follow-up and multidisciplinary treatment is essential.


Subject(s)
Collagen Type I/genetics , Ehlers-Danlos Syndrome/genetics , Hip Dislocation, Congenital/genetics , Adolescent , Adult , Child , Child, Preschool , Collagen Type I, alpha 1 Chain , Ehlers-Danlos Syndrome/epidemiology , Ehlers-Danlos Syndrome/physiopathology , Exons/genetics , Female , Genetic Predisposition to Disease , Hip Dislocation, Congenital/epidemiology , Hip Dislocation, Congenital/physiopathology , Humans , Male , Middle Aged , Mutation/genetics , Pedigree , Phenotype , Skin Abnormalities/genetics , Skin Abnormalities/physiopathology , Young Adult
12.
BMC Musculoskelet Disord ; 21(1): 6, 2020 Jan 03.
Article in English | MEDLINE | ID: mdl-31900144

ABSTRACT

BACKGROUND: Osteogenesis Imperfecta (OI) is characterized by bone fragility, and features such as blue sclerae, dentinogenesis imperfecta, hearing loss, ligamentous laxity and short stature can be present. It has long been assumed that the functional ability and quality of life of patients with OI depends primarily on the severity of skeletal deformities. However, fatigue is often mentioned in clinic by patients with all types of OI as an important modifier of their quality of life and does not always seem to be related to their functional ability. The aim of this study is to investigate whether adults with Osteogenesis Imperfecta are significantly more fatigued than the normal population. METHODS: The Fatigue Severity Scale (FSS) was distributed by mobile phone application among 151 adult patients with different OI types. Results of the FSS in the OI group were compared with two control populations from America (n = 20) and the Netherlands (n = 113). RESULTS: Ninety-nine patients (OI type 1 (n = 72), OI type 3 (n = 13), OI type 4 (n = 14) completed the FSS questionnaire. The mean FSS score of this cohort was 4.4 and significantly higher than the control populations (2.3/2.9). 65% of our cohort reported at least moderate fatigue compared with 2 control populations from America and the Netherlands. CONCLUSION: Fatigue in patients with OI is a frequently encountered problem in our expert clinic but research into this topic is sparse. This pilot study is the largest study to date investigating fatigue in patients with OI and results have been compared with two control groups. The mean FSS score of 4.4 in the OI group indicates that people with OI are generally significantly more fatigued than the control population. Further evaluation of fatigue and its influencers in a larger group of OI patients is important for future management.


Subject(s)
Cell Phone , Fatigue/diagnosis , Mobile Applications , Osteogenesis Imperfecta/diagnosis , Surveys and Questionnaires , Adolescent , Adult , Aged , Aged, 80 and over , Case-Control Studies , Cost of Illness , Cross-Sectional Studies , Fatigue/etiology , Fatigue/physiopathology , Fatigue/psychology , Female , Health Status , Humans , Male , Middle Aged , Osteogenesis Imperfecta/complications , Osteogenesis Imperfecta/physiopathology , Osteogenesis Imperfecta/psychology , Pilot Projects , Predictive Value of Tests , Quality of Life , Severity of Illness Index , Young Adult
14.
Br J Haematol ; 187(4): 509-517, 2019 11.
Article in English | MEDLINE | ID: mdl-31304589

ABSTRACT

Osteogenesis imperfecta (OI) is characterized by susceptibility to bone fractures. Other symptoms, such as easy bruising and bleeding complications during surgery necessitating transfusions, have also been reported. The aim of the cross-sectional pilot study was to assess the bleeding and bruising tendency in OI patients and to screen for possible underlying haematological disorders. Bleeding tendency was investigated using the International Society on Thrombosis and Haemostasis bleeding assessment tool (ISTH-BAT) in 22 adult OI patients. Laboratory testing was performed to investigate for bleeding disorders or abnormal coagulation. Four patients [OI type 1(n = 3), OI type 4(n = 1)] had a bleeding score (BS) fitting with a bleeding tendency, but without test results pointing to a coagulopathy. Two patients [OI type 1(n = 1), OI type 3 (n = 1)] without a bleeding tendency according to the BS had increased fibrinolysis. This is the second largest study to date addressing bleeding tendency in OI and the first study to use ISTH-BAT and elaborate laboratory testing for coagulopathies. Four patients had an increased bleeding tendency. However, laboratory testing demonstrated no bleeding disorder or abnormal coagulation. Increased fibrinolysis was demonstrated in two patients without bleeding tendency on BS. Vascular fragility as a cause of bleeding tendency in OI has been suggested earlier. Further research on bleeding tendency in OI is important.


Subject(s)
Contusions/diagnosis , Hemorrhage/diagnosis , Osteogenesis Imperfecta/pathology , Adult , Blood Coagulation Disorders , Contusions/etiology , Cross-Sectional Studies , Female , Fibrinolysis , Hemorrhage/etiology , Humans , Male , Pilot Projects , Young Adult
15.
Genet Med ; 21(9): 2081-2091, 2019 09.
Article in English | MEDLINE | ID: mdl-30837697

ABSTRACT

PURPOSE: The Ehlers-Danlos syndromes (EDS) are a group of rare inherited connective tissue disorders. Vascular EDS (vEDS) is caused by pathogenic variants in COL3A1, most frequently glycine substitutions. We describe the phenotype of the largest series of vEDS patients with glutamic acid to lysine substitutions (Glu>Lys) in COL3A1, which were all previously considered to be variants of unknown significance. METHODS: Clinical and molecular data for seven families with three different Glu>Lys substitutions in COL3A1 were analyzed. RESULTS: These Glu>Lys variants were reclassified from variants of unknown significance to either pathogenic or likely pathogenic in accordance with American College of Medical Genetics and Genomics guidelines. All individuals with these atypical variants exhibited skin hyperextensibility as seen in individuals with classical EDS and classical-like EDS and evidence of tissue fragility as seen in individuals with vEDS. CONCLUSION: The clinical data demonstrate the overlap between the different EDS subtypes and underline the importance of next-generation sequencing gene panel analysis. The three different Glu>Lys variants point toward a new variant type in COL3A1 causative of vEDS, which has consistent clinical features. This is important knowledge for COL3A1 variant interpretation. Further follow-up data are required to establish the severity of tissue fragility complications compared with patients with other recognized molecular causes of vEDS.


Subject(s)
Collagen Type III/genetics , Ehlers-Danlos Syndrome/genetics , Skin Abnormalities/genetics , Adult , Aged , Ehlers-Danlos Syndrome/classification , Ehlers-Danlos Syndrome/pathology , Female , Glutamic Acid/genetics , Glycine/genetics , High-Throughput Nucleotide Sequencing , Humans , Lysine/genetics , Male , Middle Aged , Mutation , Pedigree , Phenotype , Skin Abnormalities/pathology
16.
Genet Med ; 20(1): 42-54, 2018 01.
Article in English | MEDLINE | ID: mdl-28617417

ABSTRACT

PurposeIn 2012 we reported in six individuals a clinical condition almost indistinguishable from PLOD1-kyphoscoliotic Ehlers-Danlos syndrome (PLOD1-kEDS), caused by biallelic mutations in FKBP14, and characterized by progressive kyphoscoliosis, myopathy, and hearing loss in addition to connective tissue abnormalities such as joint hypermobility and hyperelastic skin. FKBP14 is an ER-resident protein belonging to the family of FK506-binding peptidyl-prolyl cis-trans isomerases (PPIases); it catalyzes the folding of type III collagen and interacts with type III, type VI, and type X collagens. Only nine affected individuals have been reported to date.MethodsWe report on a cohort of 17 individuals with FKBP14-kEDS and the follow-up of three previously reported patients, and provide an extensive overview of the disorder and its natural history based on clinical, biochemical, and molecular genetics data.ResultsBased on the frequency of the clinical features of 23 patients from the present and previous cohorts, we define major and minor features of FKBP14-kEDS. We show that myopathy is confirmed by histology and muscle imaging only in some patients, and that hearing impairment is predominantly sensorineural and may not be present in all individuals.ConclusionOur data further support the extensive clinical overlap with PLOD1-kEDS and show that vascular complications are rare manifestations of FKBP14-kEDS.


Subject(s)
Alleles , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/genetics , Genetic Association Studies , Mutation , Peptidylprolyl Isomerase/genetics , Phenotype , Child , Child, Preschool , Chromosome Mapping , Cohort Studies , DNA Mutational Analysis , Female , Humans , Magnetic Resonance Angiography , Magnetic Resonance Imaging , Male
17.
Hum Mutat ; 37(2): 148-54, 2016 Feb.
Article in English | MEDLINE | ID: mdl-26507355

ABSTRACT

Mandibulofacial dysostosis with microcephaly (MFDM) is a multiple malformation syndrome comprising microcephaly, craniofacial anomalies, hearing loss, dysmorphic features, and, in some cases, esophageal atresia. Haploinsufficiency of a spliceosomal GTPase, U5-116 kDa/EFTUD2, is responsible. Here, we review the molecular basis of MFDM in the 69 individuals described to date, and report mutations in 38 new individuals, bringing the total number of reported individuals to 107 individuals from 94 kindreds. Pathogenic EFTUD2 variants comprise 76 distinct mutations and seven microdeletions. Among point mutations, missense substitutions are infrequent (14 out of 76; 18%) relative to stop-gain (29 out of 76; 38%), and splicing (33 out of 76; 43%) mutations. Where known, mutation origin was de novo in 48 out of 64 individuals (75%), dominantly inherited in 12 out of 64 (19%), and due to proven germline mosaicism in four out of 64 (6%). Highly penetrant clinical features include, microcephaly, first and second arch craniofacial malformations, and hearing loss; esophageal atresia is present in an estimated ∼27%. Microcephaly is virtually universal in childhood, with some adults exhibiting late "catch-up" growth and normocephaly at maturity. Occasionally reported anomalies, include vestibular and ossicular malformations, reduced mouth opening, atrophy of cerebral white matter, structural brain malformations, and epibulbar dermoid. All reported EFTUD2 mutations can be found in the EFTUD2 mutation database (http://databases.lovd.nl/shared/genes/EFTUD2).


Subject(s)
Abnormalities, Multiple/genetics , Hearing Loss/genetics , Intellectual Disability/genetics , Mandibulofacial Dysostosis/genetics , Microcephaly/genetics , Mutation , Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/pathology , Amino Acid Motifs , Databases, Genetic , Gene Expression , Haploinsufficiency , Hearing Loss/diagnosis , Hearing Loss/pathology , Humans , Intellectual Disability/diagnosis , Intellectual Disability/pathology , Mandibulofacial Dysostosis/diagnosis , Mandibulofacial Dysostosis/pathology , Microcephaly/diagnosis , Microcephaly/pathology , Models, Molecular , Molecular Sequence Data , Penetrance , Phenotype , Protein Structure, Secondary , Protein Structure, Tertiary , RNA Splicing , Spliceosomes/genetics
19.
N Engl J Med ; 369(16): 1529-36, 2013 Oct 17.
Article in English | MEDLINE | ID: mdl-24088043

ABSTRACT

Plastin 3 (PLS3), a protein involved in the formation of filamentous actin (F-actin) bundles, appears to be important in human bone health, on the basis of pathogenic variants in PLS3 in five families with X-linked osteoporosis and osteoporotic fractures that we report here. The bone-regulatory properties of PLS3 were supported by in vivo analyses in zebrafish. Furthermore, in an additional five families (described in less detail) referred for diagnosis or ruling out of osteogenesis imperfecta type I, a rare variant (rs140121121) in PLS3 was found. This variant was also associated with a risk of fracture among elderly heterozygous women that was two times as high as that among noncarriers, which indicates that genetic variation in PLS3 is a novel etiologic factor involved in common, multi-factorial osteoporosis.


Subject(s)
Fractures, Bone/genetics , Membrane Glycoproteins/genetics , Microfilament Proteins/genetics , Osteoporosis/genetics , Adult , Animals , Bone Density/genetics , Bone Remodeling/genetics , Child , Child, Preschool , Female , Fractures, Bone/etiology , Genetic Diseases, X-Linked/genetics , Heterozygote , Humans , Male , Mutation , Osteoporosis/complications , Pedigree , Polymorphism, Single Nucleotide , Risk Factors , Young Adult , Zebrafish
20.
Hum Mutat ; 36(12): 1145-9, 2015 Dec.
Article in English | MEDLINE | ID: mdl-26247899

ABSTRACT

We report three families with arterial aneurysms and dissections in which variants predicted to be pathogenic were identified in SMAD2. Moreover, one variant occurred de novo in a proband with unaffected parents. SMAD2 is a strong candidate gene for arterial aneurysms and dissections given its role in the TGF-ß signaling pathway. Furthermore, although SMAD2 and SMAD3 probably have functionally distinct roles in cell signaling, they are structurally very similar. Our findings indicate that SMAD2 mutations are associated with arterial aneurysms and dissections and are in accordance with the observation that patients with pathogenic variants in genes encoding proteins involved in the TGF-ß signaling pathway exhibit arterial aneurysms and dissections as key features.


Subject(s)
Aneurysm/genetics , Aortic Dissection/genetics , Arteries/metabolism , Arteries/pathology , Mutation , Smad2 Protein/genetics , Adult , Alleles , Aneurysm/diagnosis , Aneurysm/metabolism , Aortic Dissection/diagnosis , Aortic Dissection/metabolism , Computational Biology/methods , Female , Genetic Association Studies , Genetic Predisposition to Disease , Genotype , Humans , Male , Middle Aged , Models, Molecular , Protein Interaction Domains and Motifs , Sequence Analysis, DNA , Smad2 Protein/chemistry , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL