Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 237
Filter
1.
Cleft Palate Craniofac J ; 59(11): 1346-1351, 2022 11.
Article in English | MEDLINE | ID: mdl-34714179

ABSTRACT

OBJECTIVE: Facial dysostosis is a group of rare craniofacial congenital disabilities requiring multidisciplinary long-term care. This report presents the phenotypic and genotypic information from South India. DESIGN: The study is a case series. SETTING: This was an international collaborative study involving a tertiary craniofacial clinic and medical genetics unit. PATIENTS, PARTICIPANTS: The participants were 9 families with 17 affected individuals of facial dysostosis. INTERVENTION: Exome analysis focused on known genes associated with acrofacial and mandibulofacial syndromes. MAIN OUTCOME MEASURE: The outcome measure was to report phenotyptic and genetic heterogeneity in affected individuals. RESULTS: A Tessier cleft was seen in 7 (41%), lower eyelid coloboma in 12 (65%), ear anomalies in 10 (59%), uniolateral or bilateral aural atresia in 4 (24%), and deafness in 6 (35%). The facial gestalt of Treacher Collins syndrome (TCS) showed extensive phenotypic variations. Pathogenic variants in TCOF1 (Treacher Collins syndrome) were seen in six families, POLR1A (acrofacial dysostosis, Cincinnati type) and EFTUD2 (mandibulofacial dysostosis with microcephaly) in one each. One family (11.1%) had no detectable variation. Five out of six probands with Treacher Collins syndrome had other affected family members (83.3%), including a non-penetrant mother, identified after sequencing. CONCLUSION: Our report illustrates the molecular heterogeneity of mandibulofacial dysostosis in India.


Subject(s)
Mandibulofacial Dysostosis , Microcephaly , Face , Genotype , Humans , Mandibulofacial Dysostosis/genetics , Microcephaly/genetics , Peptide Elongation Factors/genetics , Ribonucleoprotein, U5 Small Nuclear/genetics , Syndrome
2.
Hum Vaccin Immunother ; 17(9): 3192-3202, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34077301

ABSTRACT

Continued outbreaks of Ebola virus disease, including recent outbreaks in the Democratic Republic of the Congo (DRC), highlight the need for effective vaccine programs to combat future outbreaks. Given the population flow between DRC and Rwanda, the Rwanda Ministry of Health initiated a preventive vaccination campaign supported by a vaccination monitoring platform (VMP). The campaign aimed to vaccinate approximately 200,000 people from Rwanda's Rubavu and Rusizi districts with the two-dose vaccine regimen Ad26.ZEBOV, MVA-BN-Filo. The VMP encompassed: biometric identification (iris scanning), mobile messaging, and an interactive reporting dashboard. The VMP collected data used to register and identify participants at subsequent visits. Mobile message reminders supported compliance. To 13 November 2020, the campaign was half complete with Ad26.ZEBOV administered to 116,974 participants and MVA-BN-Filo to 76,464. MVA-BN-Filo should be given to participants approximately 8 weeks after the Ad26.ZEBOV with a compliance window of -14 and +28 days. Of the 83,850 participants who were eligible per this dosing window for the subsequent MVA-BN-Filo vaccine, 91.2% (76,453/83,850) received it and 82.9% (69,505/83,850) received it within the compliance window defined for this campaign. Utilization of the VMP was instrumental to the success of the campaign, using biometric technology, dashboard reporting of near real-time data analysis and mobile phone communication technology to support vaccine administration and monitoring. A comprehensive VMP is feasible in large-scale health-care campaigns, beneficial for public health surveillance, and can allow effective response to an infectious disease outbreak.


Subject(s)
Ebola Vaccines , Ebolavirus , Hemorrhagic Fever, Ebola , Hemorrhagic Fever, Ebola/epidemiology , Hemorrhagic Fever, Ebola/prevention & control , Humans , Immunization Programs , Rwanda/epidemiology , Vaccination
4.
Clin Genet ; 98(1): 74-79, 2020 07.
Article in English | MEDLINE | ID: mdl-32270475

ABSTRACT

Pseudoxanthoma elasticum (PXE) is a rare autosomal recessive ectopic mineralization disorder, characterized by skin, eye and cardiovascular symptoms. The most devastating ocular complication is choroidal neovascularization, which is thought to be mediated by vascular endothelial growth factor (VEGF) signaling, a molecule encoded by the VEGFA gene. As early detection and treatment is essential to preserve vision, prioritization of patients at risk is crucial, but impossible because of wide phenotypic variability and a lack of genotype-phenotype correlations for PXE. This study aimed to validate the previously suggested association of five single nucleotide VEGFA variants (rs13207351, rs833061, rs699947, rs25648 and rs1413711) with a severe PXE retinopathy in an independent cohort. Direct Sanger sequencing was performed in 100 PXE patients, with a mild (56) or severe (44) PXE retinopathy. The inclusion criteria for severe retinopathy were a unilateral best-corrected visual acuity of <5/10 and/or the need for anti-angiogenic treatment. We found a significant association of three of five variants and borderline missed significance for one. These data further suggest the VEGFA gene to be a modifier gene for the PXE retinopathy. Hereby, we provide the necessary evidence to implement these variants in ocular risk stratification and individualized patient follow-up.


Subject(s)
Genetic Markers/genetics , Polymorphism, Single Nucleotide/genetics , Pseudoxanthoma Elasticum/genetics , Retinal Diseases/genetics , Vascular Endothelial Growth Factor A/genetics , Adolescent , Adult , Aged , Aged, 80 and over , Choroidal Neovascularization/genetics , Cohort Studies , Female , Genetic Association Studies/methods , Humans , Male , Middle Aged , Prognosis , Skin/pathology , Young Adult
5.
Genet Med ; 22(1): 124-131, 2020 01.
Article in English | MEDLINE | ID: mdl-31316167

ABSTRACT

PURPOSE: Congenital contractural arachnodactyly (CCA) is an autosomal dominant connective tissue disorder manifesting joint contractures, arachnodactyly, crumpled ears, and kyphoscoliosis as main features. Due to its rarity, rather aspecific clinical presentation, and overlap with other conditions including Marfan syndrome, the diagnosis is challenging, but important for prognosis and clinical management. CCA is caused by pathogenic variants in FBN2, encoding fibrillin-2, but locus heterogeneity has been suggested. We designed a clinical scoring system and diagnostic criteria to support the diagnostic process and guide molecular genetic testing. METHODS: In this retrospective study, we assessed 167 probands referred for FBN2 analysis and classified them into a FBN2-positive (n = 44) and FBN2-negative group (n = 123) following molecular analysis. We developed a 20-point weighted clinical scoring system based on the prevalence of ten main clinical characteristics of CCA in both groups. RESULTS: The total score was significantly different between the groups (P < 0.001) and was indicative for classifying patients into unlikely CCA (total score <7) and likely CCA (total score ≥7) groups. CONCLUSIONS: Our clinical score is helpful for clinical guidance for patients suspected to have CCA, and provides a quantitative tool for phenotyping in research settings.


Subject(s)
Arachnodactyly/diagnosis , Contracture/diagnosis , Fibrillin-2/genetics , Sequence Analysis, DNA/methods , Arachnodactyly/genetics , Child , Contracture/genetics , Diagnosis, Differential , Early Diagnosis , Female , Genetic Testing , Humans , Male , Marfan Syndrome/diagnosis , Marfan Syndrome/genetics , Phenotype , Retrospective Studies , Sensitivity and Specificity
6.
Hum Mol Genet ; 28(11): 1853-1864, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30668708

ABSTRACT

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.


Subject(s)
Carboxypeptidases/genetics , Ehlers-Danlos Syndrome/genetics , Joint Instability/genetics , Repressor Proteins/genetics , Skin Abnormalities/genetics , Adolescent , Adult , Alleles , Child , Child, Preschool , Collagen/genetics , Ehlers-Danlos Syndrome/physiopathology , Extracellular Matrix/genetics , Female , Humans , Joint Instability/physiopathology , Male , Middle Aged , Mutation/genetics , Phenotype , Skin/pathology , Skin Abnormalities/physiopathology , Young Adult
7.
Hum Mol Genet ; 28(11): 1801-1809, 2019 06 01.
Article in English | MEDLINE | ID: mdl-30657919

ABSTRACT

The cyclic adenosine monophosphate 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.


Subject(s)
Cyclic AMP Response Element-Binding Protein/genetics , Endoplasmic Reticulum Stress/genetics , Nerve Tissue Proteins/genetics , Osteogenesis Imperfecta/genetics , Protein Domains/genetics , Astrocytes/metabolism , Astrocytes/pathology , COP-Coated Vesicles/genetics , Child, Preschool , Collagen Type I/chemistry , Collagen Type I/genetics , Cyclic AMP Response Element-Binding Protein/chemistry , DNA-Binding Proteins/genetics , Female , Homozygote , Humans , Male , Models, Molecular , Mutation, Missense/genetics , Nerve Tissue Proteins/chemistry , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Pedigree , Phenotype , Protein Binding , Vesicular Transport Proteins/genetics
9.
Dis Model Mech ; 11(10)2018 10 18.
Article in English | MEDLINE | ID: mdl-30355591

ABSTRACT

Targeted genome editing by CRISPR/Cas9 is extremely well fitted to generate gene disruptions, although precise sequence replacement by CRISPR/Cas9-mediated homology-directed repair (HDR) suffers from low efficiency, impeding its use for high-throughput knock-in disease modeling. In this study, we used next-generation sequencing (NGS) analysis to determine the efficiency and reliability of CRISPR/Cas9-mediated HDR using several types of single-stranded oligodeoxynucleotide (ssODN) repair templates for the introduction of disease-relevant point mutations in the zebrafish genome. Our results suggest that HDR rates are strongly determined by repair-template composition, with the most influential factor being homology-arm length. However, we found that repair using ssODNs does not only lead to precise sequence replacement but also induces integration of repair-template fragments at the Cas9 cut site. We observed that error-free repair occurs at a relatively constant rate of 1-4% when using different repair templates, which was sufficient for transmission of point mutations to the F1 generation. On the other hand, erroneous repair mainly accounts for the variability in repair rate between the different repair templates. To further improve error-free HDR rates, elucidating the mechanism behind this erroneous repair is essential. We show that the error-prone nature of ssODN-mediated repair, believed to act via synthesis-dependent strand annealing (SDSA), is most likely due to DNA synthesis errors. In conclusion, caution is warranted when using ssODNs for the generation of knock-in models or for therapeutic applications. We recommend the application of in-depth NGS analysis to examine both the efficiency and error-free nature of HDR events.This article has an associated First Person interview with the first author of the paper.


Subject(s)
CRISPR-Associated Protein 9/metabolism , CRISPR-Cas Systems/genetics , Genome , Mutation/genetics , Oligodeoxyribonucleotides/metabolism , Recombinational DNA Repair/genetics , Templates, Genetic , Zebrafish/genetics , Animals , Base Pairing , Base Sequence , DNA Breaks, Double-Stranded , Gene Knock-In Techniques , Germ Cells/metabolism , Injections
10.
Sci Rep ; 8(1): 15845, 2018 Oct 29.
Article in English | MEDLINE | ID: mdl-30374100

ABSTRACT

A correction to this article has been published and is linked from the HTML and PDF versions of this paper. The error has not been fixed in the paper.

11.
Proc Natl Acad Sci U S A ; 115(34): E8037-E8046, 2018 08 21.
Article in English | MEDLINE | ID: mdl-30082390

ABSTRACT

The type I collagenopathies are a group of heterogeneous connective tissue disorders, that are caused by mutations in the genes encoding type I collagen and include specific forms of osteogenesis imperfecta (OI) and the Ehlers-Danlos syndrome (EDS). These disorders present with a broad disease spectrum and large clinical variability of which the underlying genetic basis is still poorly understood. In this study, we systematically analyzed skeletal phenotypes in a large set of zebrafish, with diverse mutations in the genes encoding type I collagen, representing different genetic forms of human OI, and a zebrafish model resembling human EDS, which harbors a number of soft connective tissues defects, typical of EDS. Furthermore, we provide insight into how zebrafish and human type I collagen are compositionally and functionally related, which is relevant in the interpretation of human type I collagen-related disease models. Our studies reveal a high degree of intergenotype variability in phenotypic expressivity that closely correlates with associated OI severity. Furthermore, we demonstrate the potential for select mutations to give rise to phenotypic variability, mirroring the clinical variability associated with human disease pathology. Therefore, our work suggests the future potential for zebrafish to aid in identifying unknown genetic modifiers and mechanisms underlying the phenotypic variability in OI and related disorders. This will improve diagnostic strategies and enable the discovery of new targetable pathways for pharmacological intervention.


Subject(s)
Collagen Type I , Disease Models, Animal , Ehlers-Danlos Syndrome , Osteogenesis Imperfecta , Zebrafish , Animals , Animals, Genetically Modified , Collagen Type I/genetics , Collagen Type I/metabolism , Ehlers-Danlos Syndrome/genetics , Ehlers-Danlos Syndrome/metabolism , Ehlers-Danlos Syndrome/pathology , Humans , Osteogenesis Imperfecta/genetics , Osteogenesis Imperfecta/metabolism , Osteogenesis Imperfecta/pathology , Zebrafish/genetics , Zebrafish/metabolism
12.
Circ Genom Precis Med ; 11(6): e002039, 2018 06.
Article in English | MEDLINE | ID: mdl-29875124

ABSTRACT

BACKGROUND: The introduction of next-generation sequencing techniques has substantially increased the identification of new genetic variants and hence the necessity of accurate variant interpretation. In 2015, the American College of Medical Genetics and Genomics and the Association for Molecular Pathology proposed new variant interpretation guidelines. Gene-specific characteristics were, however, not considered, sometimes leading to inconsistent variant interpretation. METHODS: To allow a more uniform interpretation of variants in the FBN1 (fibrillin-1) gene, causing Marfan syndrome, we tailored these guidelines to this gene and disease. We adapted 15 of the 28 general criteria and classified 713 FBN1 variants previously identified in our laboratory as causal mutation or variant of uncertain significance according to these adapted guidelines. We then compared the agreement between previous methods and the adapted American College of Medical Genetics and Genomics and the Association for Molecular Pathology criteria. RESULTS: Agreement between the methods was 86.4% (K-alpha, 0.6). Application of the tailored guidelines resulted in an increased number of variants of uncertain significance (14.5% to 24.2%). Of the 85 variants that were downscaled to likely benign or variant of uncertain significance, 59.7% were missense variants outside a well-established functional site. Available clinical- or segregation data, necessary to further classify these types of variants, were in many cases insufficient to aid the classification. CONCLUSIONS: Our study shows that classification of variants remains challenging and may change over time. Currently, a higher level of evidence is necessary to classify a variant as pathogenic. Gene-specific guidelines may be useful to allow a more precise and uniform interpretation of the variants to accurately support clinical decision-making.


Subject(s)
Fibrillin-1/genetics , Genetics, Medical , Genomics/methods , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Mutation , Pathology, Molecular , Practice Guidelines as Topic/standards , Adult , Decision Making , Female , High-Throughput Nucleotide Sequencing , Humans , Male , Marfan Syndrome/classification , Young Adult
13.
Brain Pathol ; 28(6): 822-831, 2018 11.
Article in English | MEDLINE | ID: mdl-29722917

ABSTRACT

Ischemic stroke causes a high mortality and morbidity worldwide. It results from a complex interplay of incompletely known environmental and genetic risk factors. We investigated the ABCC6 gene as a candidate risk factor for ischemic stroke because of the increased ischemic stroke incidence in the autosomal recessive disorder pseudoxanthoma elasticum, caused by biallelic pathogenic ABCC6 variants, the higher cardiovascular risk in heterozygous carriers and the established role of ABCC6 dysfunction in myocardial ischemia. We established segregation of a known pathogenic ABCC6 variant (p.[Arg1314Gln]) in 11/19 family members of an ischemic stroke patient in a large multigenerational family suffering from ischemic stroke and/or cardiovascular disease at a relatively young age. In an independent case-control study in 424 ischemic stroke patients and 250 healthy controls, pathogenic ABCC6 variants were 4.9 times more frequent (P = 0.036; 95% CI 1.11-21.33) in the ischemic stroke patient cohort. To study cellular consequences of ABCC6 deficiency in the brain, immunostaining of brain sections in Abcc6-deficient mice and wild-type controls were performed. An upregulation of Bmp4 and Eng and a downregulation of Alk2 was identified in Abcc6-/- mice, suggesting an increase in apoptosis and angiogenesis. As both of these processes are induced in ischemia, we propose that a pro-ischemic state may explain the higher risk to suffer from ischemic stroke in patients carrying a pathogenic ABCC6 variant, as this may lower the threshold to develop acute ischemic events in these patients. In conclusion, this study identified heterozygous ABCC6 variants as a risk factor for ischemic stroke. Further, dysregulation of Bmp (Bmp4, Alk2) and Tgfß (Eng) signaling in the brain of Abcc6-/- mice could lead to a pro-ischemic state, lowering the threshold to develop acute ischemic events. These data demonstrate the importance of a molecular analysis of the ABCC6 gene in patients diagnosed with cryptogenic ischemic stroke.


Subject(s)
Multidrug Resistance-Associated Proteins/genetics , Stroke/epidemiology , Stroke/genetics , Activin Receptors, Type I/genetics , Adult , Aged , Aged, 80 and over , Animals , Apoptosis/physiology , Bone Morphogenetic Protein 4/metabolism , Case-Control Studies , Cohort Studies , Endoglin/metabolism , Female , Humans , Male , Mice , Mice, Knockout , Middle Aged , Multidrug Resistance-Associated Proteins/blood , Neovascularization, Physiologic , Pseudoxanthoma Elasticum/complications , Pseudoxanthoma Elasticum/genetics , Risk Factors , Vascular Endothelial Growth Factor A/metabolism
14.
Matrix Biol ; 70: 72-83, 2018 09.
Article in English | MEDLINE | ID: mdl-29551664

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.


Subject(s)
Amino Acid Substitution , Arteries/metabolism , Collagen Type III/genetics , Ehlers-Danlos Syndrome/genetics , Mutation , Skin/metabolism , Animals , Arteries/pathology , Collagen Type III/chemistry , Collagen Type III/deficiency , Disease Models, Animal , Ehlers-Danlos Syndrome/metabolism , Ehlers-Danlos Syndrome/mortality , Ehlers-Danlos Syndrome/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , Glycine/chemistry , Glycine/metabolism , Heterozygote , Humans , Male , Mice , Mice, Transgenic , Serine/chemistry , Serine/metabolism , Sex Factors , Skin/pathology , Tissue Culture Techniques
15.
Genet Med ; 20(10): 1236-1245, 2018 10.
Article in English | MEDLINE | ID: mdl-29323665

ABSTRACT

PURPOSE: We delineate the clinical spectrum and describe the histology in arterial tortuosity syndrome (ATS), a rare connective tissue disorder characterized by tortuosity of the large and medium-sized arteries, caused by mutations in SLC2A10. METHODS: We retrospectively characterized 40 novel ATS families (50 patients) and reviewed the 52 previously reported patients. We performed histology and electron microscopy (EM) on skin and vascular biopsies and evaluated TGF-ß signaling with immunohistochemistry for pSMAD2 and CTGF. RESULTS: Stenoses, tortuosity, and aneurysm formation are widespread occurrences. Severe but rare vascular complications include early and aggressive aortic root aneurysms, neonatal intracranial bleeding, ischemic stroke, and gastric perforation. Thus far, no reports unequivocally document vascular dissections or ruptures. Of note, diaphragmatic hernia and infant respiratory distress syndrome (IRDS) are frequently observed. Skin and vascular biopsies show fragmented elastic fibers (EF) and increased collagen deposition. EM of skin EF shows a fragmented elastin core and a peripheral mantle of microfibrils of random directionality. Skin and end-stage diseased vascular tissue do not indicate increased TGF-ß signaling. CONCLUSION: Our findings warrant attention for IRDS and diaphragmatic hernia, close monitoring of the aortic root early in life, and extensive vascular imaging afterwards. EM on skin biopsies shows disease-specific abnormalities.


Subject(s)
Arteries/abnormalities , Glucose Transport Proteins, Facilitative/genetics , Hernia, Diaphragmatic/genetics , Joint Instability/genetics , Respiratory Distress Syndrome, Newborn/genetics , Skin Diseases, Genetic/genetics , Vascular Malformations/genetics , Adolescent , Adult , Aorta/diagnostic imaging , Aorta/physiopathology , Arteries/diagnostic imaging , Arteries/physiopathology , Biopsy , Child , Child, Preschool , Connective Tissue Growth Factor/genetics , Female , Hernia, Diaphragmatic/physiopathology , Humans , Infant , Joint Instability/epidemiology , Joint Instability/physiopathology , Male , Mutation , Pedigree , Respiratory Distress Syndrome, Newborn/physiopathology , Skin/pathology , Skin Diseases, Genetic/epidemiology , Skin Diseases, Genetic/physiopathology , Smad2 Protein/genetics , Transforming Growth Factor beta/genetics , Vascular Malformations/epidemiology , Vascular Malformations/physiopathology
16.
Toxicol Sci ; 163(2): 409-419, 2018 06 01.
Article in English | MEDLINE | ID: mdl-28329870

ABSTRACT

A number of drugs can cause precipitates within renal tubules leading to crystal nephropathy. Crystal nephropathy is usually an exposure-related finding and is not uncommon in preclinical studies, where high doses are tested. An understanding of the nature of precipitates is important for human risk assessment and further development. Our aim was to investigate the ability of various imaging techniques to detect the presence of drugs or metabolites in renal crystals. We applied matrix-assisted laser desorption/ionization-Fourier transform ion cyclotron resonance mass spectrometry (MALDI-FTICR MS) imaging, Raman and infrared microspectroscopy, scanning electron microscopy coupled with energy dispersive X-ray (SEM/EDX) spectroscopy and standard histopathology to cases of drug-induced crystal nephropathy, induced in rodents and primates by 4 compounds. MALDI-FTICR MS imaging enabled the identification of the drug-related crystal content in all 4 cases of nephropathy, without reference material and with high accuracy. Crystals were composed of unchanged parent drug and/or metabolites. Similar results were obtained using Raman and infrared microspectroscopy for 2 compounds. In the absence of reference standards of metabolites, Raman and infrared microspectroscopy showed that the crystals consisted of components similar, but not identical, to the administered drug for the other compounds, a limitation for these techniques. SEM/EDX showed which counter ions were colocalized with the identified drug-related material, complementing the MALDI-FTICR MS findings. Therefore, we recommend MALDI-FTICR MS as a first-line methodology to characterize crystal nephropathies. Raman and infrared microspectroscopy may be useful when MALDI-FTICR MS imaging cannot be applied. SEM/EDX could be considered as a complementary technology.


Subject(s)
Acute Kidney Injury/diagnostic imaging , Drug-Related Side Effects and Adverse Reactions/diagnostic imaging , Kidney/drug effects , Pharmaceutical Preparations/chemistry , Animals , Crystallization , Drug Evaluation, Preclinical , Kidney/diagnostic imaging , Macaca fascicularis , Mice , Molecular Structure , Pharmaceutical Preparations/analysis , Rats , Species Specificity , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Spectrophotometry, Infrared , Spectrum Analysis, Raman
17.
Mol Genet Genomic Med ; 6(1): 15-26, 2018 01.
Article in English | MEDLINE | ID: mdl-29150909

ABSTRACT

BACKGROUND: Osteogenesis imperfecta (OI) is a heterogeneous hereditary connective tissue disorder clinically hallmarked by increased susceptibility to bone fractures. METHODS: We analyzed a cohort of 77 diagnosed OI patients from 49 unrelated Palestinian families. Next-generation sequencing technology was used to screen a panel of known OI genes. RESULTS: In 41 probands, we identified 28 different disease-causing variants of 9 different known OI genes. Eleven of the variants are novel. Ten of the 28 variants are located in COL1A1, five in COL1A2, three in BMP1, three in FKBP10, two in TMEM38B, two in P3H1, and one each in CRTAP, SERPINF1, and SERPINH1. The absence of disease-causing variants in the remaining eight probands suggests further genetic heterogeneity in OI. In general, most OI patients (90%) harbor mainly variants in type I collagen resulting in an autosomal dominant inheritance pattern. However, in our cohort almost 61% (25/41) were affected with autosomal recessive OI. Moreover, we document a 21-kb genomic deletion in the TMEM38B gene identified in 29% (12/41) of the tested probands, making it the most frequent OI-causing variant in the Palestinian population. CONCLUSION: This is the first genetic screening of an OI cohort from the Palestinian population. Our data are important for genetic counseling of OI patients and families in highly consanguineous populations.


Subject(s)
Arabs/genetics , Osteogenesis Imperfecta/genetics , Adult , Calcification, Physiologic/genetics , Collagen Type I/genetics , Consanguinity , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Family , Female , Genes, Recessive , Humans , Ion Channels/genetics , Male , Middle Aged , Mutation , Pedigree , Sequence Deletion
18.
BMC Med Ethics ; 18(1): 46, 2017 Aug 01.
Article in English | MEDLINE | ID: mdl-28764782

ABSTRACT

BACKGROUND: Carrier screening is generally performed with the aim of identifying healthy couples at risk of having a child affected with a monogenic disorder to provide them with reproductive options. Expanded carrier screening (ECS), which provides the opportunity for multiple conditions to be screened in one test, offers a more cost-effective and comprehensive option than screening for single disorders. However, implementation of ECS at a population level would have implications for genetic counseling practice. METHODS: We conducted semi-structured interviews with sixteen European clinical and molecular geneticists with expertise in carrier screening to explore their views on the implementation of ECS in the clinical setting. RESULTS: Using inductive content analysis, we identified content categories relevant to the pre- and post-test settings. Participants believed ECS would ideally be targeted at couples before pregnancy. There was some disagreement regarding the acceptability of performing ECS in individuals, with several participants actively opposing individual-based screening. In addition, participants discussed the importance of ensuring informed and voluntary participation in ECS, recommending measures to minimize external pressure on prospective parents to undergo testing. A need for adequate counseling to foster informed, autonomous reproductive decision-making and provide support for couples found to be at risk was emphasized. CONCLUSIONS: Practical challenges in optimizing pre-test education and post-test counseling should not be underestimated and they should be carefully addressed before implementing ECS in the clinical setting.


Subject(s)
Attitude of Health Personnel , Counseling , Genetic Carrier Screening/ethics , Genetic Counseling , Health Personnel , Mass Screening/ethics , Reproduction , Access to Information/ethics , Decision Making/ethics , Europe , Family Characteristics , Female , Genetic Testing , Humans , Informed Consent , Patient Selection/ethics , Pregnancy , Prospective Studies
19.
Acta Cardiol ; 72(6): 616-624, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28657492

ABSTRACT

BACKGROUND: Marfan syndrome (MFS) is a multisystemic hereditary connective tissue disease. Aortic root aneurysms and dissections are the most common and life-threatening cardiovascular disorders affecting these patients. Other cardiac manifestations include mitral valve prolapse, ventricular dysfunction and arrhythmias. Medical treatment of cardiovascular features is ultimately aimed at slowing down aortic root growth rate and preventing dissection. Losartan has been proposed as a new therapeutic tool for this purpose. To which extent losartan affects cardiac function has not been studied previously. METHODS: We designed a prospective, double-blind, randomized placebo-controlled trial to evaluate the effect of losartan added to beta-blocker therapy on aortic growth and ventricular function in patients with MFS. Secondary outcomes were aortic dissection, prophylactic aortic surgery and death. RESULTS: Twenty-two patients were enrolled in the trial. There was a mild and similar increase in the aortic root during the 3 years of follow-up in both groups (median 1 mm, IQR [-1-1.5] and 1 mm, IQR [-0.25-1] in the losartan and placebo group, respectively, p = 1). Diastolic and systolic ventricular function was normal at baseline in both groups and remained stable during the study. One patient in the placebo group presented a subclavian artery dissection during follow-up. CONCLUSION: Losartan on top of beta-blocker therapy has no additional effect on aortic growth or on cardiac function in patients with MFS. Our results are underpowered but are in line with the result from other groups. In order to have a better insight on whether a group of patients could benefit more from losartan therapy, the outcome of an on-going meta-analysis should be awaited.


Subject(s)
Aortic Aneurysm, Thoracic/prevention & control , Losartan/therapeutic use , Marfan Syndrome/drug therapy , Stroke Volume/physiology , Ventricular Dysfunction/prevention & control , Ventricular Function/drug effects , Adult , Angiotensin II Type 1 Receptor Blockers/therapeutic use , Aortic Aneurysm, Thoracic/etiology , Double-Blind Method , Echocardiography , Female , Humans , Magnetic Resonance Imaging, Cine , Male , Marfan Syndrome/complications , Middle Aged , Prospective Studies , Treatment Outcome , Ventricular Dysfunction/diagnosis , Ventricular Dysfunction/etiology , Young Adult
20.
Am J Med Genet A ; 173(4): 1047-1050, 2017 Apr.
Article in English | MEDLINE | ID: mdl-28261977

ABSTRACT

Type I collagen is the predominant protein of connective tissues such as skin and bone. Mutations in the type I collagen genes (COL1A1 and COL1A2) mainly cause osteogenesis imperfecta (OI). We describe a patient with clinical signs of Ehlers-Danlos syndrome (EDS), including fragile skin, easy bruising, recurrent luxations, and fractures resembling mild OI. Biochemical collagen analysis of the patients' dermal fibroblasts showed faint overmodification of the type I collagen bands, a finding specific for structural defects in type I collagen. Bidirectional Sanger sequencing detected an in-frame deletion in exon 44 of COL1A1 (c.3150_3158del), resulting in the deletion of three amino acids (p.Ala1053_Gly1055del) in the collagen triple helix. This COL1A1 mutation was hitherto identified in four probands with lethal OI, and never in EDS patients. As the peaks on the electropherogram corresponding to the mutant allele were decreased in intensity, we performed next generation sequencing of COL1A1 to study mosaicism in skin and blood. While approximately 9% of the reads originating from fibroblast gDNA harbored the COL1A1 deletion, the deletion was not detected in gDNA from blood. Most likely, the mild clinical symptoms observed in our patient can be explained by the mosaic state of the mutation.


Subject(s)
Collagen Type I/genetics , Ehlers-Danlos Syndrome/genetics , Mosaicism , Mutation , Osteogenesis Imperfecta/genetics , Base Sequence , Bone and Bones/metabolism , Bone and Bones/pathology , Collagen Type I, alpha 1 Chain , Ehlers-Danlos Syndrome/diagnosis , Ehlers-Danlos Syndrome/pathology , Exons , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression , High-Throughput Nucleotide Sequencing , Humans , Leukocytes, Mononuclear/cytology , Leukocytes, Mononuclear/metabolism , Organ Specificity , Osteogenesis Imperfecta/diagnosis , Osteogenesis Imperfecta/pathology , Skin/metabolism , Skin/pathology , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL
...