Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 60
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Annu Rev Genomics Hum Genet ; 23: 223-253, 2022 08 31.
Article in English | MEDLINE | ID: mdl-36044906

ABSTRACT

Genetic predisposition and risk factors such as hypertension and smoking can instigate the development of thoracic aortic aneurysm (TAA), which can lead to highly lethal aortic wall dissection and/or rupture. Monogenic defects in multiple genes involved in the elastin-contractile unit and the TGFß signaling pathway have been associated with TAA in recent years, along with several genetic modifiers and risk-conferring polymorphisms. Advances in omics technology have also provided significant insights into the processes behind aortic wall degeneration: inflammation, epigenetics, vascular smooth muscle phenotype change and depletion, reactive oxygen species generation, mitochondrial dysfunction, and angiotensin signaling dysregulation. These recent advances and findings might pave the way for a therapy that is capable of stopping and perhaps even reversing aneurysm progression.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Dissection , Aortic Dissection/genetics , Aortic Dissection/metabolism , Animals , Aortic Aneurysm, Thoracic/genetics , Aortic Aneurysm, Thoracic/metabolism , Humans , Mice , Mice, Inbred C57BL , Muscle, Smooth, Vascular/metabolism , Myocytes, Smooth Muscle/metabolism , Phenotype
2.
Am J Hum Genet ; 108(6): 1115-1125, 2021 06 03.
Article in English | MEDLINE | ID: mdl-34010605

ABSTRACT

Importin 8, encoded by IPO8, is a ubiquitously expressed member of the importin-ß protein family that translocates cargo molecules such as proteins, RNAs, and ribonucleoprotein complexes into the nucleus in a RanGTP-dependent manner. Current knowledge of the cargoes of importin 8 is limited, but TGF-ß signaling components such as SMAD1-4 have been suggested to be among them. Here, we report that bi-allelic loss-of-function variants in IPO8 cause a syndromic form of thoracic aortic aneurysm (TAA) with clinical overlap with Loeys-Dietz and Shprintzen-Goldberg syndromes. Seven individuals from six unrelated families showed a consistent phenotype with early-onset TAA, motor developmental delay, connective tissue findings, and craniofacial dysmorphic features. A C57BL/6N Ipo8 knockout mouse model recapitulates TAA development from 8-12 weeks onward in both sexes but most prominently shows ascending aorta dilatation with a propensity for dissection in males. Compliance assays suggest augmented passive stiffness of the ascending aorta in male Ipo8-/- mice throughout life. Immunohistological investigation of mutant aortic walls reveals elastic fiber disorganization and fragmentation along with a signature of increased TGF-ß signaling, as evidenced by nuclear pSmad2 accumulation. RT-qPCR assays of the aortic wall in male Ipo8-/- mice demonstrate decreased Smad6/7 and increased Mmp2 and Ccn2 (Ctgf) expression, reinforcing a role for dysregulation of the TGF-ß signaling pathway in TAA development. Because importin 8 is the most downstream TGF-ß-related effector implicated in TAA pathogenesis so far, it offers opportunities for future mechanistic studies and represents a candidate drug target for TAA.


Subject(s)
Aortic Aneurysm, Thoracic/etiology , Loss of Function Mutation , Loss of Heterozygosity , Phenotype , beta Karyopherins/genetics , Adult , Animals , Aortic Aneurysm, Thoracic/metabolism , Aortic Aneurysm, Thoracic/pathology , Child , Child, Preschool , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Pedigree , Signal Transduction , Syndrome , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism , Young Adult , beta Karyopherins/metabolism
3.
Int J Mol Sci ; 25(9)2024 May 04.
Article in English | MEDLINE | ID: mdl-38732244

ABSTRACT

Cardiovascular outcome in Marfan syndrome (MFS) patients most prominently depends on aortic aneurysm progression with subsequent aortic dissection. Angiotensin II receptor blockers (ARBs) prevent aneurysm formation in MFS mouse models. In patients, ARBs only slow down aortic dilation. Downstream signalling from the angiotensin II type 1 receptor (AT1R) is mediated by G proteins and ß-arrestin recruitment. AT1R also interacts with the monocyte chemoattractant protein-1 (MCP-1) receptor, resulting in inflammation. In this study, we explore the targeting of ß-arrestin signalling in MFS mice by administering TRV027. Furthermore, because high doses of the ARB losartan, which has been proven beneficial in MFS, cannot be achieved in humans, we investigate a potential additive effect by combining lower concentrations of losartan (25 mg/kg/day and 5 mg/kg/day) with barbadin, a ß-arrestin blocker, and DMX20, a C-C chemokine receptor type 2 (CCR2) blocker. A high dose of losartan (50 mg/kg/day) slowed down aneurysm progression compared to untreated MFS mice (1.73 ± 0.12 vs. 1.96 ± 0.08 mm, p = 0.0033). TRV027, the combination of barbadin with losartan (25 mg/kg/day), and DMX-200 (90 mg/kg/day) with a low dose of losartan (5 mg/kg/day) did not show a significant beneficial effect. Our results confirm that while losartan effectively halts aneurysm formation in Fbn1C1041G/+ MFS mice, neither TRV027 alone nor any of the other compounds combined with lower doses of losartan demonstrate a notable impact on aneurysm advancement. It appears that complete blockade of AT1R function, achieved by administrating a high dosage of losartan, may be necessary for inhibiting aneurysm progression in MFS.


Subject(s)
Angiotensin II Type 1 Receptor Blockers , Aortic Aneurysm , Losartan , Marfan Syndrome , Signal Transduction , Mice , Marfan Syndrome/drug therapy , Marfan Syndrome/pathology , Disease Models, Animal , Aortic Aneurysm/drug therapy , Aortic Aneurysm/prevention & control , Oligopeptides/administration & dosage , Aorta, Thoracic/drug effects , Aorta, Thoracic/pathology , Pyrimidines/administration & dosage , Drug Combinations , Losartan/administration & dosage , Angiotensin Receptor Antagonists/administration & dosage , Signal Transduction/drug effects , Humans , Angiotensin II Type 1 Receptor Blockers/administration & dosage
4.
Hum Mutat ; 43(7): 815-831, 2022 07.
Article in English | MEDLINE | ID: mdl-35419902

ABSTRACT

Different pathogenic variants in the fibrillin-1 gene (FBN1) cause Marfan syndrome and acromelic dysplasias. Whereas the musculoskeletal features of Marfan syndrome involve tall stature, arachnodactyly, joint hypermobility, and muscle hypoplasia, acromelic dysplasia patients present with short stature, brachydactyly, stiff joints, and hypermuscularity. Similarly, pathogenic variants in the fibrillin-2 gene (FBN2) cause either a Marfanoid congenital contractural arachnodactyly or a FBN2-related acromelic dysplasia that most prominently presents with brachydactyly. The phenotypic and molecular resemblances between both the FBN1 and FBN2-related disorders suggest that reciprocal pathomechanistic lessons can be learned. In this review, we provide an updated overview and comparison of the phenotypic and mutational spectra of both the "tall" and "short" fibrillinopathies. The future parallel functional study of both FBN1/2-related disorders will reveal new insights into how pathogenic fibrillin variants differently affect the fibrillin microfibril network and/or growth factor homeostasis in clinically opposite syndromes. This knowledge may eventually be translated into new therapeutic approaches by targeting or modulating the fibrillin microfibril network and/or the signaling pathways under its control.


Subject(s)
Fibrillin-1 , Fibrillin-2 , Marfan Syndrome , Musculoskeletal Abnormalities/genetics , Brachydactyly , Fibrillin-1/genetics , Fibrillin-2/genetics , Humans , Marfan Syndrome/genetics , Phenotype
5.
Hum Mutat ; 43(12): 1824-1828, 2022 12.
Article in English | MEDLINE | ID: mdl-35819173

ABSTRACT

Pathogenic variants in JAG1 are known to cause Alagille syndrome (ALGS), a disorder that primarily affects the liver, lung, kidney, and skeleton. Whereas cardiac symptoms are also frequently observed in ALGS, thoracic aortic aneurysms have only been reported sporadically in postmortem autopsies. We here report two families with segregating JAG1 variants that present with isolated aneurysmal disease, as well as the first histological evaluation of aortic aneurysm tissue of a JAG1 variant carrier. Our observations shed more light on the pathomechanisms behind aneurysm formation in JAG1 variant harboring individuals and underline the importance of cardiovascular imaging in the clinical follow-up of such individuals.


Subject(s)
Alagille Syndrome , Humans , Jagged-1 Protein/genetics , Jagged-1 Protein/metabolism , Alagille Syndrome/genetics , Heart , Calcium-Binding Proteins
6.
Am J Physiol Cell Physiol ; 322(6): C1214-C1222, 2022 06 01.
Article in English | MEDLINE | ID: mdl-35476501

ABSTRACT

The class I small leucine-rich proteoglycan biglycan is a crucial structural extracellular matrix component that interacts with a wide range of extracellular matrix molecules. In addition, biglycan is involved in sequestering growth factors such as transforming growth factor-ß and bone morphogenetic proteins and thereby regulating pathway activity. Biglycan consists of a 42-kDa core protein linked to two glycosaminoglycan side chains and both are involved in protein interactions. Biglycan is encoded by the BGN gene located on the X-chromosome and is expressed in various tissues, including vascular tissue, skin, brain, kidney, lung, the immune system, and the musculoskeletal system. Although an increasing amount of data on the biological function of biglycan in the vasculature has been produced, its role in thoracic aortic aneurysms is still not fully elucidated. This review focuses on the role of biglycan in the healthy thoracic aorta and the development of thoracic aortic aneurysm and dissections in both mice and humans.


Subject(s)
Aorta, Thoracic , Transforming Growth Factor beta , Animals , Aorta, Thoracic/metabolism , Biglycan/genetics , Biglycan/metabolism , Bone Morphogenetic Proteins , Extracellular Matrix Proteins/genetics , Mice , Signal Transduction , Transforming Growth Factor beta/genetics , Transforming Growth Factor beta/metabolism
7.
Genet Med ; 24(7): 1583-1591, 2022 07.
Article in English | MEDLINE | ID: mdl-35499524

ABSTRACT

PURPOSE: CTR9 is a subunit of the PAF1 complex (PAF1C) that plays a crucial role in transcription regulation by binding CTR9 to RNA polymerase II. It is involved in transcription-coupled histone modification through promoting H3K4 and H3K36 methylation. We describe the clinical and molecular studies in 13 probands, harboring likely pathogenic CTR9 missense variants, collected through GeneMatcher. METHODS: Exome sequencing was performed in all individuals. CTR9 variants were assessed through 3-dimensional modeling of the activated human transcription complex Pol II-DSIF-PAF-SPT6 and the PAF1/CTR9 complex. H3K4/H3K36 methylation analysis, mitophagy assessment based on tetramethylrhodamine ethyl ester perchlorate immunofluorescence, and RNA-sequencing in skin fibroblasts from 4 patients was performed. RESULTS: Common clinical findings were variable degrees of intellectual disability, hypotonia, joint hyperlaxity, speech delay, coordination problems, tremor, and autism spectrum disorder. Mild dysmorphism and cardiac anomalies were less frequent. For 11 CTR9 variants, de novo occurrence was shown. Three-dimensional modeling predicted a likely disruptive effect of the variants on local CTR9 structure and protein interaction. Additional studies in fibroblasts did not unveil the downstream functional consequences of the identified variants. CONCLUSION: We describe a neurodevelopmental disorder caused by (mainly) de novo variants in CTR9, likely affecting PAF1C function.


Subject(s)
Autism Spectrum Disorder , Intellectual Disability , Neurodevelopmental Disorders , Phosphoproteins , Transcription Factors , Gene Expression Regulation , Heterozygote , Humans , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Phosphoproteins/genetics , Transcription Factors/genetics
8.
Genet Med ; 24(5): 1045-1053, 2022 05.
Article in English | MEDLINE | ID: mdl-35058154

ABSTRACT

PURPOSE: In a large cohort of 373 pediatric patients with Marfan syndrome (MFS) with a severe cardiovascular phenotype, we explored the proportion of patients with MFS with a pathogenic FBN1 variant and analyzed whether the type/location of FBN1 variants was associated with specific clinical characteristics and response to treatment. Patients were recruited on the basis of the following criteria: aortic root z-score > 3, age 6 months to 25 years, no prior or planned surgery, and aortic root diameter < 5 cm. METHODS: Targeted resequencing and deletion/duplication testing of FBN1 and related genes were performed. RESULTS: We identified (likely) pathogenic FBN1 variants in 91% of patients. Ectopia lentis was more frequent in patients with dominant-negative (DN) variants (61%) than in those with haploinsufficient variants (27%). For DN FBN1 variants, the prevalence of ectopia lentis was highest in the N-terminal region (84%) and lowest in the C-terminal region (17%). The association with a more severe cardiovascular phenotype was not restricted to DN variants in the neonatal FBN1 region (exon 25-33) but was also seen in the variants in exons 26 to 49. No difference in the therapeutic response was detected between genotypes. CONCLUSION: Important novel genotype-phenotype associations involving both cardiovascular and extra-cardiovascular manifestations were identified, and existing ones were confirmed. These findings have implications for prognostic counseling of families with MFS.


Subject(s)
Ectopia Lentis , Marfan Syndrome , Biological Variation, Population , Child , Ectopia Lentis/complications , Ectopia Lentis/genetics , Fibrillin-1/genetics , Fibrillins/genetics , Genotype , Humans , Marfan Syndrome/genetics , Mutation , Phenotype
9.
Am J Hum Genet ; 103(2): 288-295, 2018 08 02.
Article in English | MEDLINE | ID: mdl-30032985

ABSTRACT

The natriuretic peptide signaling pathway has been implicated in many cellular processes, including endochondral ossification and bone growth. More precisely, different mutations in the NPR-B receptor and the CNP ligand have been identified in individuals with either short or tall stature. In this study we show that the NPR-C receptor (encoded by NPR3) is also important for the regulation of linear bone growth. We report four individuals, originating from three different families, with a phenotype characterized by tall stature, long digits, and extra epiphyses in the hands and feet. In addition, aortic dilatation was observed in two of these families. In each affected individual, we identified a bi-allelic loss-of-function mutation in NPR3. The missense mutations (c.442T>C [p.Ser148Pro] and c.1088A>T [p.Asp363Val]) resulted in intracellular retention of the NPR-C receptor and absent localization on the plasma membrane, whereas the nonsense mutation (c.1524delC [p.Tyr508∗]) resulted in nonsense-mediated mRNA decay. Biochemical analysis of plasma from two affected and unrelated individuals revealed a reduced NTproNP/NP ratio for all ligands and also high cGMP levels. These data strongly suggest a reduced clearance of natriuretic peptides by the defective NPR-C receptor and consequently increased activity of the NPR-A/B receptors. In conclusion, this study demonstrates that loss-of-function mutations in NPR3 result in increased NPR-A/B signaling activity and cause a phenotype marked by enhanced bone growth and cardiovascular abnormalities.


Subject(s)
Connective Tissue/abnormalities , Loss of Heterozygosity/genetics , Mutation/genetics , Natriuretic Peptide, C-Type/genetics , Adolescent , Bone Development/genetics , Cardiovascular Abnormalities/genetics , Child , Cyclic GMP/genetics , Female , Humans , Male , Signal Transduction/genetics
10.
Adv Exp Med Biol ; 1348: 265-272, 2021.
Article in English | MEDLINE | ID: mdl-34807424

ABSTRACT

Meester-Loeys syndrome is an X-linked form of syndromic thoracic aortic aneurysm, characterized by the involvement of multiple organ systems. More specifically, the cardiovascular, skeletal, craniofacial, cutaneous and neurological systems are affected. Clear clinical overlap with Marfan syndrome and Loeys-Dietz syndrome is observed. Aortic dissections occur typically at young ages and are most often observed in males. Meester-Loeys syndrome is caused by loss-of-function mutations in BGN, encoding the small leucine-rich proteoglycan biglycan. Although functional consequences of these mutations remain largely elusive, increased TGF-ß signaling has been observed. Novel insights will provide opportunities for preventive therapeutic interventions.


Subject(s)
Aortic Aneurysm, Thoracic , Aortic Dissection , Loeys-Dietz Syndrome , Marfan Syndrome , Humans , Loeys-Dietz Syndrome/diagnosis , Loeys-Dietz Syndrome/genetics , Male , Mutation
11.
Int J Mol Sci ; 22(13)2021 Jul 01.
Article in English | MEDLINE | ID: mdl-34281165

ABSTRACT

Thoracic aortic aneurysm and dissection (TAAD) is a major cause of cardiovascular morbidity and mortality. Loss-of-function variants in LOX, encoding the extracellular matrix crosslinking enzyme lysyl oxidase, have been reported to cause familial TAAD. Using a next-generation TAAD gene panel, we identified five additional probands carrying LOX variants, including two missense variants affecting highly conserved amino acids in the LOX catalytic domain and three truncating variants. Connective tissue manifestations are apparent in a substantial fraction of the variant carriers. Some LOX variant carriers presented with TAAD early in life, while others had normal aortic diameters at an advanced age. Finally, we identified the first patient with spontaneous coronary artery dissection carrying a LOX variant. In conclusion, our data demonstrate that loss-of-function LOX variants cause a spectrum of aortic and arterial aneurysmal disease, often combined with connective tissue findings.


Subject(s)
Aortic Aneurysm, Thoracic/genetics , Protein-Lysine 6-Oxidase/genetics , Adult , Aortic Dissection/genetics , Aortic Dissection/physiopathology , Aorta/metabolism , Aortic Aneurysm, Thoracic/physiopathology , Arteries/metabolism , Connective Tissue/metabolism , Connective Tissue Diseases/genetics , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Middle Aged , Mutation/genetics , Pedigree , Protein-Lysine 6-Oxidase/metabolism
12.
Acta Neuropathol ; 139(6): 1001-1024, 2020 06.
Article in English | MEDLINE | ID: mdl-32172343

ABSTRACT

Parkinson's disease (PD) is a progressive neurodegenerative brain disease presenting with a variety of motor and non-motor symptoms, loss of midbrain dopaminergic neurons in the substantia nigra pars compacta and the occurrence of α-synuclein-positive Lewy bodies in surviving neurons. Here, we performed whole exome sequencing in 52 early-onset PD patients and identified 3 carriers of compound heterozygous mutations in the ATP10B P4-type ATPase gene. Genetic screening of a Belgian PD and dementia with Lewy bodies (DLB) cohort identified 4 additional compound heterozygous mutation carriers (6/617 PD patients, 0.97%; 1/226 DLB patients, 0.44%). We established that ATP10B encodes a late endo-lysosomal lipid flippase that translocates the lipids glucosylceramide (GluCer) and phosphatidylcholine (PC) towards the cytosolic membrane leaflet. The PD associated ATP10B mutants are catalytically inactive and fail to provide cellular protection against the environmental PD risk factors rotenone and manganese. In isolated cortical neurons, loss of ATP10B leads to general lysosomal dysfunction and cell death. Impaired lysosomal functionality and integrity is well known to be implicated in PD pathology and linked to multiple causal PD genes and genetic risk factors. Our results indicate that recessive loss of function mutations in ATP10B increase risk for PD by disturbed lysosomal export of GluCer and PC. Both ATP10B and glucocerebrosidase 1, encoded by the PD risk gene GBA1, reduce lysosomal GluCer levels, emerging lysosomal GluCer accumulation as a potential PD driver.


Subject(s)
Adenosine Triphosphatases/genetics , Glucosylceramides/metabolism , Lysosomes/metabolism , Membrane Transport Proteins/genetics , Mutation/genetics , Parkinson Disease/genetics , Aged , Aged, 80 and over , Dopaminergic Neurons/metabolism , Female , Glucosylceramidase/genetics , Glucosylceramides/genetics , Humans , Lewy Bodies/pathology , Lysosomes/genetics , Male , Middle Aged , Parkinson Disease/metabolism , Parkinson Disease/pathology , alpha-Synuclein/metabolism
13.
J Med Genet ; 56(4): 220-227, 2019 04.
Article in English | MEDLINE | ID: mdl-29967133

ABSTRACT

BACKGROUND: Missense variants in SMAD2, encoding a key transcriptional regulator of transforming growth factor beta signalling, were recently reported to cause arterial aneurysmal disease. OBJECTIVES: The aims of the study were to identify the genetic disease cause in families with aortic/arterial aneurysmal disease and to further define SMAD2 genotype-phenotype correlations. METHODS AND RESULTS: Using gene panel sequencing, we identified a SMAD2 nonsense variant and four SMAD2 missense variants, all affecting highly conserved amino acids in the MH2 domain. The premature stop codon (c.612dup; p.(Asn205*)) was identified in a marfanoid patient with aortic root dilatation and in his affected father. A p.(Asn318Lys) missense variant was found in a Marfan syndrome (MFS)-like case who presented with aortic root aneurysm and in her affected daughter with marfanoid features and mild aortic dilatation. In a man clinically diagnosed with Loeys-Dietz syndrome (LDS) that presents with aortic root dilatation and marked tortuosity of the neck vessels, another missense variant, p.(Ser397Tyr), was identified. This variant was also found in his affected daughter with hypertelorism and arterial tortuosity, as well as his affected mother. The third missense variant, p.(Asn361Thr), was discovered in a man presenting with coronary artery dissection. Variant genotyping in three unaffected family members confirmed its absence. The last missense variant, p.(Ser467Leu), was identified in a man with significant cardiovascular and connective tissue involvement. CONCLUSION: Taken together, our data suggest that heterozygous loss-of-function SMAD2 variants can cause a wide spectrum of autosomal dominant aortic and arterial aneurysmal disease, combined with connective tissue findings reminiscent of MFS and LDS.


Subject(s)
Aneurysm/etiology , Aortic Dissection/etiology , Aortic Dissection/pathology , Arteries/pathology , Genetic Variation , Smad2 Protein/genetics , Adult , Aged , Alleles , Amino Acid Substitution , Aneurysm/pathology , Child , Facies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Male , Marfan Syndrome/complications , Marfan Syndrome/genetics , Middle Aged , Mutation , Pedigree , Phenotype , Smad2 Protein/metabolism
14.
Connect Tissue Res ; 60(2): 146-154, 2019 03.
Article in English | MEDLINE | ID: mdl-29732924

ABSTRACT

Marfan syndrome (MFS) is a multi-systemic autosomal dominant condition caused by mutations in the gene (FBN1) coding for fibrillin-1. Mutations have been associated with a wide range of overlapping phenotypes. Here, we report on an extended family presenting with skeletal, ocular and cardiovascular clinical features. The 37-year-old male propositus, who had chest pain, dyspnea and shortness of breath, was first diagnosed based on the revised Ghent criteria and then subjected to molecular genetic analyses. FBN1 sequencing of the proband as well as available affected family members revealed the presence of a novel variant, c.7828G>C (p.Glu2610Gln), which was not present in any of the unaffected family members. In silico analyses demonstrated that the Glu2610 residue is part of the conserved DINE motif found at the beginning of each cbEGF domain of FBN1. The substitution of Glu2610 with Gln decreased fibrillin-1 production accordingly. Despite the fact that this variation appears to be primarily responsible for the etiology of MFS in the present family, our findings suggest that variable clinical expressions of the disease phenotype should be considered critically by the physicians.


Subject(s)
Fibrillin-1/genetics , Marfan Syndrome/genetics , Marfan Syndrome/pathology , Mutation/genetics , Adolescent , Adult , Amino Acid Sequence , Base Sequence , Child , Computer Simulation , Family , Female , Fibrillin-1/chemistry , Heterozygote , Humans , Male , Pedigree , Phenotype
16.
Hum Mutat ; 39(5): 621-634, 2018 05.
Article in English | MEDLINE | ID: mdl-29392890

ABSTRACT

The Loeys-Dietz syndrome (LDS) is a connective tissue disorder affecting the cardiovascular, skeletal, and ocular system. Most typically, LDS patients present with aortic aneurysms and arterial tortuosity, hypertelorism, and bifid/broad uvula or cleft palate. Initially, mutations in transforming growth factor-ß (TGF-ß) receptors (TGFBR1 and TGFBR2) were described to cause LDS, hereby leading to impaired TGF-ß signaling. More recently, TGF-ß ligands, TGFB2 and TGFB3, as well as intracellular downstream effectors of the TGF-ß pathway, SMAD2 and SMAD3, were shown to be involved in LDS. This emphasizes the role of disturbed TGF-ß signaling in LDS pathogenesis. Since most literature so far has focused on TGFBR1/2, we provide a comprehensive review on the known and some novel TGFB2/3 and SMAD2/3 mutations. For TGFB2 and SMAD3, the clinical manifestations, both of the patients previously described in the literature and our newly reported patients, are summarized in detail. This clearly indicates that LDS concerns a disorder with a broad phenotypical spectrum that is still emerging as more patients will be identified. All mutations described here are present in the corresponding Leiden Open Variant Database.


Subject(s)
Genetic Association Studies , Loeys-Dietz Syndrome/genetics , Mutation/genetics , Smad2 Protein/genetics , Smad3 Protein/genetics , Transforming Growth Factor beta2/genetics , Transforming Growth Factor beta3/genetics , Animals , Disease Models, Animal , Humans , Loeys-Dietz Syndrome/diagnosis , Mice , Signal Transduction/genetics
17.
Trends Genet ; 31(3): 140-9, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25703649

ABSTRACT

Parkinson disease (PD) and Parkinson-plus syndromes are genetically heterogeneous neurological diseases. Initial studies into the genetic causes of PD relied on classical molecular genetic approaches in well-documented case families. More recently, these approaches have been combined with exome sequencing and together have identified 15 causal genes. Additionally, genome-wide association studies (GWASs) have discovered over 25 genetic risk factors. Elucidation of the genetic architecture of sporadic and familial parkinsonism, however, has lagged behind that of simple Mendelian conditions, suggesting the existence of features confounding genetic data interpretation. Here we discuss the successes and potential pitfalls of gene discovery in PD and related disorders in the post-genomic era. With an estimated 30% of trait variance currently unexplained, tackling current limitations will further expedite gene discovery and lead to increased application of these genetic insights in molecular diagnostics using gene panel and exome sequencing strategies.


Subject(s)
Exome/genetics , Genome, Human/genetics , Genomics , Parkinson Disease/genetics , Genetic Predisposition to Disease , Genome-Wide Association Study , Humans , Mutation , Parkinson Disease/therapy , Risk Factors , Sequence Analysis, DNA
18.
BMC Med Genet ; 19(1): 140, 2018 08 08.
Article in English | MEDLINE | ID: mdl-30089473

ABSTRACT

BACKGROUND: Mutations in the X-linked gene filamin A (FLNA), encoding the actin-binding protein FLNA, cause a wide spectrum of connective tissue, skeletal, cardiovascular and/or gastrointestinal manifestations. Males are typically more severely affected than females with common pre- or perinatal death. CASE PRESENTATION: We provide a genotype- and phenotype-oriented literature overview of FLNA hemizygous mutations and report on two live-born male FLNA mutation carriers. Firstly, we identified a de novo, missense mutation (c.238C > G, p.(Leu80Val)) in a five-year old Indian boy who presented with periventricular nodular heterotopia, increased skin laxity, joint hypermobility, mitral valve prolapse with regurgitation and marked facial features (e.g. a flat face, orbital fullness, upslanting palpebral fissures and low-set ears). Secondly, we identified two cis-located FLNA mutations (c.7921C > G, p.(Pro2641Ala); c.7923delC, p.(Tyr2642Thrfs*63)) in a Bosnian patient with Ehlers-Danlos syndrome-like features such as skin translucency and joint hypermobility. This patient also presented with brain anomalies, pectus excavatum, mitral valve prolapse, pulmonary hypertension and dilatation of the pulmonary arteries. He died from heart failure in his second year of life. CONCLUSIONS: These two new cases expand the list of live-born FLNA mutation-positive males with connective tissue disease from eight to ten, contributing to a better knowledge of the genetic and phenotypic spectrum of FLNA-related disease.


Subject(s)
Connective Tissue Diseases/genetics , Filamins/metabolism , Mutation/genetics , Adolescent , Adult , Child , Child, Preschool , Connective Tissue/metabolism , Ehlers-Danlos Syndrome/genetics , Genes, X-Linked/genetics , Genotype , Humans , Infant, Newborn , Male , Middle Aged , Pedigree , Phenotype , Young Adult
19.
Genet Med ; 19(4): 386-395, 2017 04.
Article in English | MEDLINE | ID: mdl-27632686

ABSTRACT

PURPOSE: Thoracic aortic aneurysm and dissection (TAAD) is typically inherited in an autosomal dominant manner, but rare X-linked families have been described. So far, the only known X-linked gene is FLNA, which is associated with the periventricular nodular heterotopia type of Ehlers-Danlos syndrome. However, mutations in this gene explain only a small number of X-linked TAAD families. METHODS: We performed targeted resequencing of 368 candidate genes in a cohort of 11 molecularly unexplained Marfan probands. Subsequently, Sanger sequencing of BGN in 360 male and 155 female molecularly unexplained TAAD probands was performed. RESULTS: We found five individuals with loss-of-function mutations in BGN encoding the small leucine-rich proteoglycan biglycan. The clinical phenotype is characterized by early-onset aortic aneurysm and dissection. Other recurrent findings include hypertelorism, pectus deformity, joint hypermobility, contractures, and mild skeletal dysplasia. Fluorescent staining revealed an increase in TGF-ß signaling, evidenced by an increase in nuclear pSMAD2 in the aortic wall. Our results are in line with those of prior reports demonstrating that Bgn-deficient male BALB/cA mice die from aortic rupture. CONCLUSION: In conclusion, BGN gene defects in humans cause an X-linked syndromic form of severe TAAD that is associated with preservation of elastic fibers and increased TGF-ß signaling.Genet Med 19 4, 386-395.


Subject(s)
Aortic Aneurysm, Thoracic/genetics , Aortic Dissection/genetics , Biglycan/genetics , Mutation , Aortic Dissection/metabolism , Aortic Aneurysm, Thoracic/metabolism , Biglycan/metabolism , Cells, Cultured , Female , Genes, X-Linked , Genetic Predisposition to Disease , Humans , Male , Pedigree , Sequence Analysis, DNA/methods , Signal Transduction , Transforming Growth Factor beta/metabolism
20.
Hum Mutat ; 37(6): 524-31, 2016 06.
Article in English | MEDLINE | ID: mdl-26919284

ABSTRACT

Marfan syndrome (MFS) is a rare, autosomal-dominant, multisystem disorder, presenting with skeletal, ocular, skin, and cardiovascular symptoms. Significant clinical overlap with other systemic connective tissue diseases, including Loeys-Dietz syndrome (LDS), Shprintzen-Goldberg syndrome (SGS), and the MASS phenotype, has been documented. In MFS and LDS, the cardiovascular manifestations account for the major cause of patient morbidity and mortality, rendering them the main target for therapeutic intervention. Over the past decades, gene identification studies confidently linked the aforementioned syndromes, as well as nonsyndromic aneurysmal disease, to genetic defects in proteins related to the transforming growth factor (TGF)-ß pathway, greatly expanding our knowledge on the disease mechanisms and providing us with novel therapeutic targets. As a result, the focus of the developing pharmacological treatment strategies is shifting from hemodynamic stress management to TGF-ß antagonism. In this review, we discuss the insights that have been gained in the molecular biology of MFS and related disorders over the past 25 years.


Subject(s)
Arachnodactyly/genetics , Craniosynostoses/genetics , Loeys-Dietz Syndrome/genetics , Marfan Syndrome/genetics , Mitral Valve Prolapse/genetics , Myopia/genetics , Skin Diseases/genetics , Transforming Growth Factor beta/genetics , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Animals , Arachnodactyly/drug therapy , Craniosynostoses/drug therapy , Gene Expression Regulation , Gene Regulatory Networks/drug effects , Genetic Predisposition to Disease , Humans , Loeys-Dietz Syndrome/drug therapy , Marfan Syndrome/drug therapy , Mitral Valve Prolapse/drug therapy , Myopia/drug therapy , Signal Transduction/drug effects , Skin Diseases/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL