Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Lipids Health Dis ; 23(1): 93, 2024 Apr 01.
Article in English | MEDLINE | ID: mdl-38561799

ABSTRACT

BACKGROUND: Circulating ceramide (Cer) drives various pathological processes associated with cardiovascular diseases, liver illness, and diabetes mellitus. Although recognized as predictors of cardiometabolic diseases (CMD) in research and clinical settings, their potential for predicting CMD risk in individuals under 18 remains unexplored. OBJECTIVES: This study was designed to utilize Liquid Chromatography-Mass Spectrometry (LC-MS/MS) methodology to determine the biological reference ranges for Cer in plasma samples of Emirati children and develop a risk assessment score (CERT-1) based on Cer concentrations. METHODS: Using LC-MS/MS, we developed a method to measure five Cer species in plasma samples of 582 Emirati participants aged 5-17. We used the circulating concentrations of these Cer to determine their reference intervals in this population. We employed traditional statistical analyses to develop a risk score (CERT-1) and assess the association between Cer levels and conventional biomarkers of CMD. RESULTS: We validated a high-throughput methodology using LC-MS/MS to quantify five Cer species in human plasma. Reference values for this population (n = 582) were quantified: CerC16:0 (0.12-0.29 µmol/L), CerC18:0 (0.019-0.067 µmol/L), CerC22:0 (0.102-0.525 µmol/L), CerC24:0 (0.65-1.54 µmol/L) and CerC24:1 (0.212-0.945 µmol/L). We devised a risk assessment score (CERT-1) based on plasma Cer content in the study participants, showing that 72.5% have low to moderate risk and 9.3% are at a higher risk of developing CMD. Our analyses also revealed a significant correlation (P < 0.05) between this score and the conventional risk factors linked to CMD, indicating its potential clinical implication. CONCLUSION: This study presents a clinical-scaled LC-MS/MS methodology for assessing clinically relevant Cer, setting reference ranges, and developing a risk score (CERT-1) for young Emirati individuals. Our findings can enhance primary risk prediction and inform the management and follow-up of CMD from an early age.


Subject(s)
Cardiometabolic Risk Factors , Ceramides , Child , Humans , Adolescent , Chromatography, Liquid/methods , United Arab Emirates/epidemiology , Tandem Mass Spectrometry/methods
2.
Int J Mol Sci ; 24(11)2023 May 23.
Article in English | MEDLINE | ID: mdl-37298070

ABSTRACT

Cardiovascular disease (CVD) during pregnancy varies significantly worldwide, influenced by factors such as access to healthcare, delayed diagnosis, causes, and risk factors. Our study sought to explore the spectrum of CVD present in pregnant women in the United Arab Emirates to better understand this population's unique needs and challenges. Central to our study is an emphasis on the importance of implementing a multidisciplinary approach that involves the collaboration of obstetricians, cardiologists, geneticists, and other healthcare professionals to ensure that patients receive comprehensive and coordinated care. This approach can also help identify high-risk patients and implement preventive measures to reduce the occurrence of adverse maternal outcomes. Furthermore, increasing awareness among women about the risk of CVD during pregnancy and obtaining detailed family histories can help in the early identification and management of these conditions. Genetic testing and family screening can also aid in identifying inherited CVD that can be passed down through families. To illustrate the significance of such an approach, we provide a comprehensive analysis of five women's cases from our retrospective study of 800 women. The findings from our study emphasize the importance of addressing maternal cardiac health in pregnancy and the need for targeted interventions and improvements in the existing healthcare system to reduce adverse maternal outcomes.


Subject(s)
Cardiomyopathies , Cardiovascular Diseases , Obstetrics , Humans , Pregnancy , Female , Retrospective Studies , Risk Assessment , Cardiovascular Diseases/diagnosis , Cardiovascular Diseases/genetics
3.
Genome Res ; 29(7): 1047-1056, 2019 07.
Article in English | MEDLINE | ID: mdl-31227601

ABSTRACT

Approximately 2% of de novo single-nucleotide variants (SNVs) appear as part of clustered mutations that create multinucleotide variants (MNVs). MNVs are an important source of genomic variability as they are more likely to alter an encoded protein than a SNV, which has important implications in disease as well as evolution. Previous studies of MNVs have focused on their mutational origins and have not systematically evaluated their functional impact and contribution to disease. We identified 69,940 MNVs and 91 de novo MNVs in 6688 exome-sequenced parent-offspring trios from the Deciphering Developmental Disorders Study comprising families with severe developmental disorders. We replicated the previously described MNV mutational signatures associated with DNA polymerase zeta, an error-prone translesion polymerase, and the APOBEC family of DNA deaminases. We estimate the simultaneous MNV germline mutation rate to be 1.78 × 10-10 mutations per base pair per generation. We found that most MNVs within a single codon create a missense change that could not have been created by a SNV. MNV-induced missense changes were, on average, more physicochemically divergent, were more depleted in highly constrained genes (pLI ≥ 0.9), and were under stronger purifying selection compared with SNV-induced missense changes. We found that de novo MNVs were significantly enriched in genes previously associated with developmental disorders in affected children. This shows that MNVs can be more damaging than SNVs even when both induce missense changes, and are an important variant type to consider in relation to human disease.


Subject(s)
Developmental Disabilities/genetics , Exome , Mutation , Child , DNA Mutational Analysis , Humans , Mutation Rate , Mutation, Missense , Nucleotides , Polymorphism, Single Nucleotide
4.
Clin Genet ; 101(4): 403-410, 2022 04.
Article in English | MEDLINE | ID: mdl-34988996

ABSTRACT

Here, we delineate the phenotype of two siblings with a bi-allelic frameshift variant in MMP15 gene with congenital cardiac defects, cholestasis, and dysmorphism. Genome sequencing analysis revealed a recently reported homozygous frameshift variant (c.1058delC, p.Pro353Glnfs*102) in MMP15 gene that co-segregates with the phenotype in the family in a recessive mode of inheritance. Relative quantification of MMP15 mRNA showed evidence of degradation of the mutated transcript, presumably by nonsense mediated decay. Likewise, MMP15: p.Gly231Arg, a concurrently reported homozygous missense variant in another patient exhibiting a similar phenotype, was predicted to disrupt zinc ion binding to the MMP-15 enzyme catalytic domain, which is essential for substrate proteolysis, by structural modeling. Previous animal models and cellular findings suggested that MMP15 plays a crucial role in the formation of endocardial cushions. These findings confirm that MMP15 is an important gene in human development, particularly cardiac, and that its loss of function is likely to cause a severe disorder phenotype.


Subject(s)
Cholestasis , Heart Defects, Congenital , Jaundice , Matrix Metalloproteinase 15/genetics , Animals , Failure to Thrive/genetics , Heart Defects, Congenital/genetics , Homozygote , Humans , Phenotype
5.
Eur Heart J ; 42(48): 4947-4960, 2021 12 21.
Article in English | MEDLINE | ID: mdl-34293101

ABSTRACT

AIMS: Recent clinical trials indicate that sodium-glucose cotransporter 2 (SGLT2) inhibitors improve cardiovascular outcomes in heart failure patients, but the underlying mechanisms remain unknown. We explored the direct effects of canagliflozin, an SGLT2 inhibitor with mild SGLT1 inhibitory effects, on myocardial redox signalling in humans. METHODS AND RESULTS: Study 1 included 364 patients undergoing cardiac surgery. Right atrial appendage biopsies were harvested to quantify superoxide (O2.-) sources and the expression of inflammation, fibrosis, and myocardial stretch genes. In Study 2, atrial tissue from 51 patients was used ex vivo to study the direct effects of canagliflozin on NADPH oxidase activity and nitric oxide synthase (NOS) uncoupling. Differentiated H9C2 and primary human cardiomyocytes (hCM) were used to further characterize the underlying mechanisms (Study 3). SGLT1 was abundantly expressed in human atrial tissue and hCM, contrary to SGLT2. Myocardial SGLT1 expression was positively associated with O2.- production and pro-fibrotic, pro-inflammatory, and wall stretch gene expression. Canagliflozin reduced NADPH oxidase activity via AMP kinase (AMPK)/Rac1signalling and improved NOS coupling via increased tetrahydrobiopterin bioavailability ex vivo and in vitro. These were attenuated by knocking down SGLT1 in hCM. Canagliflozin had striking ex vivo transcriptomic effects on myocardial redox signalling, suppressing apoptotic and inflammatory pathways in hCM. CONCLUSIONS: We demonstrate for the first time that canagliflozin suppresses myocardial NADPH oxidase activity and improves NOS coupling via SGLT1/AMPK/Rac1 signalling, leading to global anti-inflammatory and anti-apoptotic effects in the human myocardium. These findings reveal a novel mechanism contributing to the beneficial cardiac effects of canagliflozin.


Subject(s)
Canagliflozin , Sodium-Glucose Transporter 2 Inhibitors , Canagliflozin/metabolism , Canagliflozin/pharmacology , Humans , Myocardium , Myocytes, Cardiac/metabolism , Oxidation-Reduction , Sodium-Glucose Transporter 2 Inhibitors/pharmacology
6.
Am J Hum Genet ; 99(3): 683-694, 2016 09 01.
Article in English | MEDLINE | ID: mdl-27545674

ABSTRACT

The ubiquitin fold modifier 1 (UFM1) cascade is a recently identified evolutionarily conserved ubiquitin-like modification system whose function and link to human disease have remained largely uncharacterized. By using exome sequencing in Finnish individuals with severe epileptic syndromes, we identified pathogenic compound heterozygous variants in UBA5, encoding an activating enzyme for UFM1, in two unrelated families. Two additional individuals with biallelic UBA5 variants were identified from the UK-based Deciphering Developmental Disorders study and one from the Northern Finland Intellectual Disability cohort. The affected individuals (n = 9) presented in early infancy with severe irritability, followed by dystonia and stagnation of development. Furthermore, the majority of individuals display postnatal microcephaly and epilepsy and develop spasticity. The affected individuals were compound heterozygous for a missense substitution, c.1111G>A (p.Ala371Thr; allele frequency of 0.28% in Europeans), and a nonsense variant or c.164G>A that encodes an amino acid substitution p.Arg55His, but also affects splicing by facilitating exon 2 skipping, thus also being in effect a loss-of-function allele. Using an in vitro thioester formation assay and cellular analyses, we show that the p.Ala371Thr variant is hypomorphic with attenuated ability to transfer the activated UFM1 to UFC1. Finally, we show that the CNS-specific knockout of Ufm1 in mice causes neonatal death accompanied by microcephaly and apoptosis in specific neurons, further suggesting that the UFM1 system is essential for CNS development and function. Taken together, our data imply that the combination of a hypomorphic p.Ala371Thr variant in trans with a loss-of-function allele in UBA5 underlies a severe infantile-onset encephalopathy.


Subject(s)
Alleles , Brain Diseases/genetics , Brain Diseases/metabolism , Mutation/genetics , Proteins/genetics , Ubiquitin-Activating Enzymes/genetics , Ubiquitin/metabolism , Animals , Animals, Newborn , Apoptosis , Brain Diseases/pathology , Central Nervous System/metabolism , Central Nervous System/pathology , Cohort Studies , Epilepsy/genetics , Exome/genetics , Exons/genetics , Fibroblasts/metabolism , Fibroblasts/pathology , Finland , Gene Frequency , Heterozygote , Humans , Infant , Intellectual Disability/genetics , Mice , Mice, Knockout , Microcephaly/genetics , Microcephaly/pathology , Neurons/metabolism , Neurons/pathology , Proteins/metabolism , Spasms, Infantile/genetics , Spasms, Infantile/metabolism
7.
Int J Obes (Lond) ; 43(12): 2536-2544, 2019 12.
Article in English | MEDLINE | ID: mdl-30464235

ABSTRACT

OBJECTIVES: Very low calorie diets (VLCDs) are effective at clearing hepatic steatosis and improving insulin sensitivity. Whilst long-term weight loss is beneficial to the cardiovascular system, the acute elevation in fatty acids during caloric restriction is potentially detrimental to cardiac metabolism and function. We sought to investigate any cardiovascular changes occurring over the course of a modern VLCD regime, alongside the expected peripheral metabolic improvements. METHODS: 25 obese volunteers (BMI 36.8 ± 5.8 kg/m2) underwent magnetic resonance imaging, echocardiography, metabolic profiling, and bio-impedance analysis before 1 and 8 weeks following a VLCD (800 kcal/day). Results were compared to 15 age- and sex-matched controls. RESULTS: After 1 week of VLCD, despite only modest weight loss, significant drops occurred in liver fat and insulin resistance (HOMA-IR; by 14-50%, all p < 0.01). In contrast, myocardial triglyceride content (MTGC) increased (by 48%, p = 0.030), and was associated with deterioration in both systolic (LVEF by 4%, p = 0.041) and diastolic function (e/e' 8.6 ± 1.4 to 9.4 ± 1.7, p = 0.019). Aortic stiffness also increased by 35% (p = 0.015). At 8 weeks, liver steatosis and visceral fat were lower than baseline (by 20-55%, p < 0.001), and peripheral metabolic improvements continued. MTGC also fell to below baseline (1.5 ± 0.6 vs 2.1 ± 1%, p = 0.05) with improved myocardial function (e/e' 8.6 ± 1.4 to 7.5 ± 1.5, p = 0.003). CONCLUSIONS: Whilst VLCDs result in dramatic improvements in insulin resistance, they are associated with transient but significant cardiovascular functional decline, which may have an impact on those with the coexisting cardiac disease. However, after 8 weeks, the diet was associated with normalisation of cardiac function, suggesting they may form a potential therapeutic intervention for diastolic dysfunction in obesity and diabetes.


Subject(s)
Caloric Restriction/adverse effects , Cardiomyopathies , Obesity/diet therapy , Ventricular Dysfunction , Adult , Blood Pressure/physiology , Cardiomyopathies/etiology , Cardiomyopathies/physiopathology , Fatty Liver/physiopathology , Female , Humans , Intra-Abdominal Fat/physiology , Male , Middle Aged , Triglycerides/blood , Ventricular Dysfunction/etiology , Ventricular Dysfunction/physiopathology , Weight Loss
8.
J Med Genet ; 55(2): 122-130, 2018 02.
Article in English | MEDLINE | ID: mdl-29122926

ABSTRACT

BACKGROUND: Bone dysplasias are a large group of disorders affecting the growth and structure of the skeletal system. METHODS: In the present study, we report the clinical and molecular delineation of a new form of syndromic autosomal recessive spondylometaphyseal dysplasia (SMD) in two Emirati first cousins. They displayed postnatal growth deficiency causing profound limb shortening with proximal and distal segments involvement, narrow chest, radiological abnormalities involving the spine, pelvis and metaphyses, corneal clouding and intellectual disability. Whole genome homozygosity mapping localised the genetic cause to 11q12.1-q13.1, a region spanning 19.32 Mb with ~490 genes. Using whole exome sequencing, we identified four novel homozygous variants within the shared block of homozygosity. Pathogenic variants in genes involved in phospholipid metabolism, such as PLCB4 and PCYT1A, are known to cause bone dysplasia with or without eye anomalies, which led us to select PLCB3 as a strong candidate. This gene encodes phospholipase C ß 3, an enzyme that converts phosphatidylinositol 4,5 bisphosphate (PIP2) to inositol 1,4,5 triphosphate (IP3) and diacylglycerol. RESULTS: The identified variant (c.2632G>T) substitutes a serine for a highly conserved alanine within the Ha2' element of the proximal C-terminal domain. This disrupts binding of the Ha2' element to the catalytic core and destabilises PLCB3. Here we show that this hypomorphic variant leads to elevated levels of PIP2 in patient fibroblasts, causing disorganisation of the F-actin cytoskeleton. CONCLUSIONS: Our results connect a homozygous loss of function variant in PLCB3 with a new SMD associated with corneal dystrophy and developmental delay (SMDCD).


Subject(s)
Corneal Dystrophies, Hereditary/genetics , Osteochondrodysplasias/genetics , Phosphatidylinositols/metabolism , Phospholipase C beta/genetics , Amino Acid Substitution , Child , Child, Preschool , Chromosomes, Human, Pair 11 , Corneal Dystrophies, Hereditary/etiology , Developmental Disabilities/etiology , Developmental Disabilities/genetics , Female , Homozygote , Humans , Infant, Newborn , Intellectual Disability/genetics , Male , Osteochondrodysplasias/etiology , Pedigree , Phosphatidylinositols/genetics , Phospholipase C beta/metabolism , Signal Transduction/genetics
9.
Hum Mol Genet ; 23(13): 3456-66, 2014 Jul 01.
Article in English | MEDLINE | ID: mdl-24501276

ABSTRACT

Whereas many genes associated with intellectual disability (ID) encode synaptic proteins, transcriptional defects leading to ID are less well understood. We studied a large, consanguineous pedigree of Arab origin with seven members affected with ID and mild dysmorphic features. Homozygosity mapping and linkage analysis identified a candidate region on chromosome 17 with a maximum multipoint logarithm of odds score of 6.01. Targeted high-throughput sequencing of the exons in the candidate region identified a homozygous 4-bp deletion (c.169_172delCACT) in the METTL23 (methyltransferase like 23) gene, which is predicted to result in a frameshift and premature truncation (p.His57Valfs*11). Overexpressed METTL23 protein localized to both nucleus and cytoplasm, and physically interacted with GABPA (GA-binding protein transcription factor, alpha subunit). GABP, of which GABPA is a component, is known to regulate the expression of genes such as THPO (thrombopoietin) and ATP5B (ATP synthase, H+ transporting, mitochondrial F1 complex, beta polypeptide) and is implicated in a wide variety of important cellular functions. Overexpression of METTL23 resulted in increased transcriptional activity at the THPO promoter, whereas knockdown of METTL23 with siRNA resulted in decreased expression of ATP5B, thus revealing the importance of METTL23 as a regulator of GABPA function. The METTL23 mutation highlights a new transcriptional pathway underlying human intellectual function.


Subject(s)
DNA Modification Methylases/metabolism , GA-Binding Protein Transcription Factor/metabolism , Cell Nucleus/metabolism , Cytoplasm/metabolism , DNA Modification Methylases/genetics , Female , GA-Binding Protein Transcription Factor/genetics , Genotype , Humans , Immunoprecipitation , Male , Mitochondrial Proton-Translocating ATPases/genetics , Mitochondrial Proton-Translocating ATPases/metabolism , Polymorphism, Single Nucleotide/genetics , Protein Binding , RNA, Small Interfering , Thrombopoietin/genetics , Thrombopoietin/metabolism , Two-Hybrid System Techniques
10.
Am J Med Genet A ; 170(8): 2111-8, 2016 08.
Article in English | MEDLINE | ID: mdl-27183861

ABSTRACT

Clinical classification of overgrowth syndromes represents a challenge since a wide spectrum of disorders result in marked overgrowth. Therefore, there is a continuous effort to identify the genetic basis of these disorders that will eventually facilitate their molecular classification. Here, we have identified the genetic etiology and the pathogenetic mechanism underlying a rare autosomal recessive overgrowth syndrome in three affected siblings. The overgrowth phenotype in the patients was accompanied by developmental delay, learning disabilities, and variable congenital abnormalities. To elucidate the genetic etiology of the disorder, whole-genome genotyping and whole-exome sequencing were used. The disease was mapped to 3p21.1-p14.2 and 11q13.1-q13.4, where an in-frame insertion (c.175_176insTAA) in FIBP gene was revealed. The resulting indel (p.H59LN) was predicted to change the protein conformation with likely deleterious effect on its function as one of the fibroblast growth factor signaling mediators. In vitro cellular proliferation assay and in situ hypridization in vivo were then performed to understand the pathophysiology of the disease. The patients' skin fibroblasts showed an increased proliferation capacity compared to the controls' explaining the observed overgrowth phenotype. In addition, we detected Fibp expression most notably in the brains of mice embryos suggesting a possible effect on cognitive functions early in development. To date, only one patient has been reported with a homozygous nonsense mutation in FIBP exhibiting an overgrowth syndrome with multiple congenital abnormalities. Taken all together, these findings provide convincing evidence implicating FIBP aberrations in the newly recognized overgrowth syndrome and expand the associated phenotypes to include possible Wilms tumor predisposition. © 2016 Wiley Periodicals, Inc.


Subject(s)
Carrier Proteins/genetics , Genes, Recessive , Growth Disorders/genetics , Intellectual Disability/genetics , Kidney/abnormalities , Membrane Proteins/genetics , Mutation , Wilms Tumor/etiology , Adolescent , Animals , Cell Proliferation , Child , Child, Preschool , Chromosome Mapping , DNA Mutational Analysis , Exome , Female , Gene Expression , Gene Expression Regulation, Developmental , Genetic Association Studies , Genotype , Growth Disorders/diagnosis , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Intellectual Disability/diagnosis , Male , Mice , Mice, Transgenic , Pedigree , Phenotype , Syndrome , Wilms Tumor/diagnosis
11.
Am J Med Genet A ; 170A(1): 156-61, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26395437

ABSTRACT

The gene encoding the AT-rich interaction domain-containing protein 1B (ARID1B) has recently been shown to be one of the most frequently mutated genes in patients with intellectual disability (ID). The phenotypic spectrums associated with variants in this gene vary widely ranging for mild to severe non-specific ID to Coffin-Siris syndrome. In this study, we evaluated three children from a consanguineous Emirati family affected with ID and dysmorphic features. Genomic DNA from all affected siblings was analyzed using CGH array and whole-exome sequencing (WES). Based on a recessive mode of inheritance, homozygous or compound heterozygous variants shared among all three affected children could not be identified. However, further analysis revealed a heterozygous variant (c.4318C>T; p.Q1440*) in the three affected children in an autosomal dominant ID causing gene, ARID1B. This variant was absent in peripheral blood samples obtained from both parents and unaffected siblings. Therefore, we propose that the most likely explanation for this situation is that one of the parents is a gonadal mosaic for the variant. To the best of our knowledge, this is the first report of a gonadal mosaicism inheritance of an ARID1B variant leading to familial ID recurrence.


Subject(s)
Abnormalities, Multiple/genetics , DNA-Binding Proteins/genetics , Exome/genetics , Face/abnormalities , Hand Deformities, Congenital/genetics , Intellectual Disability/genetics , Micrognathism/genetics , Mosaicism , Mutation/genetics , Neck/abnormalities , Transcription Factors/genetics , Abnormalities, Multiple/pathology , Adolescent , Child , Face/pathology , Female , Hand Deformities, Congenital/pathology , Heterozygote , Humans , Intellectual Disability/pathology , Male , Micrognathism/pathology , Neck/pathology , Pedigree , Siblings
12.
Hum Mutat ; 34(3): 498-505, 2013 Mar.
Article in English | MEDLINE | ID: mdl-23255084

ABSTRACT

We have recently shown that the hemorrhagic destruction of the brain, subependymal, calcification, and congenital cataracts is caused by biallelic mutations in the gene encoding junctional adhesion molecule 3 (JAM3) protein. Affected members from three new families underwent detailed clinical examination including imaging of the brain. Affected individuals presented with a distinctive phenotype comprising hemorrhagic destruction of the brain, subependymal calcification, and congenital cataracts. All patients had a catastrophic clinical course resulting in death. Sequencing the coding exons of JAM3 revealed three novel homozygous mutations: c.2T>G (p.M1R), c.346G>A (p.E116K), and c.656G>A (p.C219Y). The p.M1R mutation affects the start codon and therefore is predicted to impair protein synthesis. Cellular studies showed that the p.C219Y mutation resulted in a significant retention of the mutated protein in the endoplasmic reticulum, suggesting a trafficking defect. The p.E116K mutant traffics normally to the plasma membrane as the wild-type and may have lost its function due to the lack of interaction with an interacting partner. Our data further support the importance of JAM3 in the development and function of the vascular system and the brain.


Subject(s)
Brain/pathology , Calcinosis/genetics , Cataract/congenital , Cataract/genetics , Cell Adhesion Molecules/genetics , Cerebral Hemorrhage/genetics , Australia , Cell Adhesion Molecules/deficiency , Exons , Female , HeLa Cells , Homozygote , Humans , Infant, Newborn , Intensive Care Units, Neonatal , Male , Microscopy, Confocal , Mutation, Missense , Pedigree , Phenotype , Sequence Analysis, DNA , Spain , Turkey , United Arab Emirates
13.
Am J Hum Genet ; 87(6): 882-9, 2010 Dec 10.
Article in English | MEDLINE | ID: mdl-21109224

ABSTRACT

The tight junction, or zonula occludens, is a specialized cell-cell junction that regulates epithelial and endothelial permeability, and it is an essential component of the blood-brain barrier in the cerebrovascular endothelium. In addition to functioning as a diffusion barrier, tight junctions are also involved in signal transduction. In this study, we identified a homozygous mutation in the tight-junction protein gene JAM3 in a large consanguineous family from the United Arab Emirates. Some members of this family had a rare autosomal-recessive syndrome characterized by severe hemorrhagic destruction of the brain, subependymal calcification, and congenital cataracts. Their clinical presentation overlaps with some reported cases of pseudo-TORCH syndrome as well as with cases involving mutations in occludin, another component of the tight-junction complex. However, massive intracranial hemorrhage distinguishes these patients from others. Homozygosity mapping identified the disease locus in this family on chromosome 11q25 with a maximum multipoint LOD score of 6.15. Sequence analysis of genes in the candidate interval uncovered a mutation in the canonical splice-donor site of intron 5 of JAM3. RT-PCR analysis of a patient lymphoblast cell line confirmed abnormal splicing, leading to a frameshift mutation with early termination. JAM3 is known to be present in vascular endothelium, although its roles in cerebral vasculature have not been implicated. Our results suggest that JAM3 is essential for maintaining the integrity of the cerebrovascular endothelium as well as for normal lens development in humans.


Subject(s)
Calcinosis/genetics , Cataract/congenital , Cell Adhesion Molecules/genetics , Cerebral Hemorrhage/genetics , Ependyma/pathology , Homozygote , Mutation , Tight Junctions/metabolism , Cataract/genetics , Child , Female , Humans , Infant , Infant, Newborn , Male , Pedigree
14.
Mol Cell Biochem ; 373(1-2): 247-57, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23124896

ABSTRACT

Hereditary haemorrhagic telangiectasia (HHT) is an autosomal dominant disease characterised by vascular dysplasia and increased bleeding that affect 1 in 5,000 people world-wide. Pathology is linked to mutations in genes encoding components of the heteromeric transforming growth factor-beta receptor (TGF-beta) and SMAD signalling pathway. Indeed HHT1 and HHT2 result from mutations in the genes encoding endoglin and activin-like kinase 1 (ALK1), TGF-beta receptor components. However, the fundamental cellular defects underlying HHT is poorly understood. Previously using confocal microscopy and N-glycosylation analysis, we found evidence that defective trafficking of endoglin from the endoplasmic reticulum (ER) to the plasma membrane is a mechanism underlying HHT1 in some patients. In this study, we used confocal microscopy to investigate whether a similar mechanism contributes to HHT2 pathology. To do this we expressed wild-type ALK1 and a number of HHT2 patient mutant variants as C-terminally tagged EGFP fusion proteins and tested their localisation in HeLa cells. We found that wild-type ALK1-EGFP was targeted predominantly to the plasma membrane, as evidenced by its colocalisation with the co-expressed HA-tagged endoglin. However, we found that in the majority of cases analysed the HHT2 patient mutant protein was retained within the ER as indicated by their colocalisation with the ER resident marker (calnexin) and lack of colocalisation with cell surface associated HA-endoglin. We conclude that defective trafficking and retention in the ER of mutant ALK1 protein is a possible mechanism of HHT2 in some patients.


Subject(s)
Activin Receptors, Type II/genetics , Activin Receptors, Type II/metabolism , Endoplasmic Reticulum/enzymology , Telangiectasia, Hereditary Hemorrhagic/genetics , Calnexin/metabolism , Green Fluorescent Proteins/genetics , Green Fluorescent Proteins/metabolism , HeLa Cells , Humans , Microscopy, Fluorescence , Mutation, Missense , Protein Transport , Recombinant Fusion Proteins/genetics , Recombinant Fusion Proteins/metabolism , Telangiectasia, Hereditary Hemorrhagic/enzymology
15.
Birth Defects Res A Clin Mol Teratol ; 97(7): 456-62, 2013 Jul.
Article in English | MEDLINE | ID: mdl-23696134

ABSTRACT

We report on a Palestinian family with three affected individuals exhibiting progeroid syndrome characterized by intrauterine growth retardation, a progeroid appearance, failure to thrive, short stature, and hypotonia. The progeroid features were evident at birth. All the affected members of this family have survived beyond the neonatal period and one of them is currently a 27-year-old adult. As parental consanguinity suggested an autosomal recessive mode of inheritance, we employed homozygosity mapping using single nucleotide polymorphism arrays followed by next generation whole exome sequencing to identify the disease-causing gene. We were able to identify a single block of homozygosity shared between all the affected members of the studied family spanning 2.3 Mb on chromosome 19p13.3p13.2. However, Sanger sequencing of known genes and whole exome sequencing of the three affected sibs did not reveal a convincing causal mutation. These findings are anticipated to open the way for the identification of the molecular causes underlying this syndrome.


Subject(s)
Chromosomes, Human, Pair 19/genetics , Exome , Family , Fetal Growth Retardation/genetics , Homozygote , Pedigree , Progeria/genetics , Adult , Chromosome Mapping/methods , DNA Mutational Analysis , Female , Fetal Growth Retardation/mortality , Follow-Up Studies , Humans , Male , Progeria/mortality , Survival Rate
16.
J Am Coll Cardiol ; 82(4): 317-332, 2023 07 25.
Article in English | MEDLINE | ID: mdl-37468187

ABSTRACT

BACKGROUND: Visceral obesity is directly linked to increased cardiovascular risk, including heart failure. OBJECTIVES: This study explored the ability of human epicardial adipose tissue (EAT)-derived microRNAs (miRNAs) to regulate the myocardial redox state and clinical outcomes. METHODS: This study screened for miRNAs expressed and released from human EAT and tested for correlations with the redox state in the adjacent myocardium in paired EAT/atrial biopsy specimens from patients undergoing cardiac surgery. Three miRNAs were then tested for causality in an in vitro model of cardiomyocytes. At a clinical level, causality/directionality were tested using genome-wide association screening, and the underlying mechanisms were explored using human biopsy specimens, as well as overexpression of the candidate miRNAs and their targets in vitro and in vivo using a transgenic mouse model. The final prognostic value of the discovered targets was tested in patients undergoing cardiac surgery, followed up for a median of 8 years. RESULTS: EAT miR-92a-3p was related to lower oxidative stress in human myocardium, a finding confirmed by using genetic regulators of miR-92a-3p in the human heart and EAT. miR-92a-3p reduced nicotinamide adenine dinucleotide phosphate (NADPH)-oxidase-derived superoxide (O2.-) by targeting myocardial expression of WNT5A, which regulated Rac1-dependent activation of NADPH oxidases. Finally, high miR-92a-3p levels in EAT were independently related with lower risk of adverse cardiovascular events. CONCLUSIONS: EAT-derived miRNAs exert paracrine effects on the human heart. Indeed miR-92a-3p suppresses the wingless-type MMTV integration site family, member 5a/Rac1/NADPH oxidase axis and improves the myocardial redox state. EAT-derived miR-92a-3p is related to improved clinical outcomes and is a rational therapeutic target for the prevention and treatment of obesity-related heart disease.


Subject(s)
Genome-Wide Association Study , MicroRNAs , Humans , Mice , Animals , MicroRNAs/genetics , MicroRNAs/metabolism , Myocardium/metabolism , Oxidation-Reduction , Mice, Transgenic , Adipose Tissue/metabolism
17.
Hum Mol Genet ; 19(11): 2239-50, 2010 Jun 01.
Article in English | MEDLINE | ID: mdl-20223752

ABSTRACT

Spondylo-meta-epiphyseal dysplasia (SMED) with short limbs and abnormal calcifications (SMED-SL) is a rare, autosomal recessive human growth disorder, characterized by disproportionate short stature, short limbs, short broad fingers, abnormal metaphyses and epiphyses, platyspondyly and premature calcifications. Recently, three missense mutations and one splice-site mutation in the DDR2 gene were identified as causative genetic defects for SMED-SL, but the underlying cellular and biochemical mechanisms were not explored. Here we report a novel DDR2 missense mutation, c.337G>A (p.E113K), that causes SMED-SL in two siblings in the United Arab Emirates. Another DDR2 missense mutation, c.2254C>T (p.R752C), matching one of the previously reported SMED-SL mutations, was found in a second affected family. DDR2 is a plasma membrane receptor tyrosine kinase that functions as a collagen receptor. We expressed DDR2 constructs with the identified point mutations in human cell lines and evaluated their localization and functional properties. We found that all SMED-SL missense mutants were defective in collagen-induced receptor activation and that the three previously reported mutants (p.T713I, p.I726R and p.R752C) were retained in the endoplasmic reticulum. The novel mutant (p.E113K), in contrast, trafficked normally, like wild-type DDR2, but failed to bind collagen. This finding is in agreement with our recent structural data identifying Glu113 as an important amino acid in the DDR2 ligand-binding site. Our data thus demonstrate that SMED-SL can result from at least two different loss-of-function mechanisms: namely defects in DDR2 targeting to the plasma membrane or the loss of its ligand-binding activity.


Subject(s)
Models, Molecular , Osteochondrodysplasias/genetics , Osteochondrodysplasias/metabolism , Receptor Protein-Tyrosine Kinases/genetics , Receptor Protein-Tyrosine Kinases/metabolism , Receptors, Mitogen/genetics , Receptors, Mitogen/metabolism , Base Sequence , Child , DNA Primers/genetics , Discoidin Domain Receptors , Endoplasmic Reticulum/metabolism , HeLa Cells , Humans , Immunohistochemistry , Male , Molecular Sequence Data , Mutation, Missense/genetics , Osteochondrodysplasias/diagnostic imaging , Pedigree , Protein Binding/genetics , Protein Transport/genetics , Protein Transport/physiology , Radiography , Receptor Protein-Tyrosine Kinases/chemistry , Receptors, Collagen/metabolism , Receptors, Mitogen/chemistry , Sequence Analysis, DNA , Signal Transduction/genetics , Signal Transduction/physiology , Transfection , United Arab Emirates
18.
Birth Defects Res A Clin Mol Teratol ; 94(7): 553-6, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22678705

ABSTRACT

BACKGROUND: Camptodactyly-arthropathy-coxa vara-pericarditis syndrome (CACP) is a clinically heterogenous congenital disorder caused by mutations in proteoglycan 4 (PRG4), a chondroitin sulfate proteoglycan that acts as a lubricant for the cartilage surface. Although CACP is a rare genetic disorder, several cases were described in the literature from ethnically different populations including Caucasian, Egyptian, Saudi Arabian, Pakistani, and Korean. We report CACP for the first time in United Arab Emirates. METHODS: Direct sequencing of all the coding exons and splice sites of the PRG4 gene was performed for all the members of the affected family. RESULTS: The studied family is consanguineous and has multiple affected members from different branches showing congenital camptodactyly with arthropathy, the hallmarks of CACP. All the affected family members lack pericarditis, but one of them was born with cataract, which has never been documented in any of the previously reported cases of CACP. Molecular analysis revealed a novel homozygous insertion of a cytosine nucleotide (c.1320dupC) in the highly repetitive portion of the coding sequence of the PRG4 gene. The detected mutation caused a frameshift in the cDNA sequence and created a premature termination codon (p.P440fsX197), which is likely to result in a nonfunctional protein. CONCLUSION: We report a family from the United Arab Emirates with typical features of CACP in whom one of the children had in addition, a bilateral congenital cataract. We also report the identification of a novel null mutation in PRG4 confirming the genetic homogeneity of CACP.


Subject(s)
Arthropathy, Neurogenic/genetics , Cataract/congenital , Coxa Vara/genetics , Hand Deformities, Congenital/genetics , Mutation , Phenotype , Proteoglycans/genetics , Synovitis/genetics , Adolescent , Child , Child, Preschool , Codon , Consanguinity , Humans , Male , Pedigree , United Arab Emirates
19.
Front Mol Biosci ; 9: 828199, 2022.
Article in English | MEDLINE | ID: mdl-35281255

ABSTRACT

Endoglin, also known as cluster of differentiation 105 (CD105), is an auxiliary receptor in the TGFß signaling pathway. It is predominantly expressed in endothelial cells as a component of the heterotetrameric receptor dimers comprising type I, type II receptors and the binding ligands. Mutations in the gene encoding Endoglin (ENG) have been associated with hereditary hemorrhagic telangiectasia type 1 (HHT1), an autosomal dominant inherited disease that is generally characterized by vascular malformation. Secretory and many endomembrane proteins synthesized in the Endoplasmic reticulum (ER) are subjected to stringent quality control mechanisms to ensure that only properly folded and assembled proteins are trafficked forward through the secretory pathway to their sites of action. We have previously demonstrated that some Endoglin variants causing HHT1 are trapped in the ER and fail to traffic to their normal localization in plasma membrane, which suggested the possible involvement of ER associated protein degradation (ERAD) in their molecular pathology. In this study, we have investigated, for the first time, the degradation routes of Endoglin wild type and two mutant variants, P165L and V105D, and previously shown to be retained in the ER. Stably transfected HEK293 cells were treated with proteasomal and lysosomal inhibitors in order to elucidate the exact molecular mechanisms underlying the loss of function phenotype associated with these variants. Our results have shown that wild type Endoglin has a relatively short half-life of less than 2 hours and degrades through both the lysosomal and proteasomal pathways, whereas the two mutant disease-causing variants show high stability and predominantly degrades through the proteasomal pathway. Furthermore, we have demonstrated that Endoglin variants P165L and V105D are significantly accumulated in HEK293 cells deficient in HRD1 E3 ubiquitin ligase; a major ERAD component. These results implicate the ERAD mechanism in the pathology of HHT1 caused by the two variants. It is expected that these results will pave the way for more in-depth research studies that could provide new windows for future therapeutic interventions.

20.
Front Genet ; 13: 1053999, 2022.
Article in English | MEDLINE | ID: mdl-36583020

ABSTRACT

We reported a 22-year-old Emirati male with autosomal recessive primary hypertrophic osteoarthropathy caused by a possibly pathogenic homozygous non-synonymous variant in the SLCO2A1 gene (NM_005630.3: c.289C>T, p. Arg97Cys) presenting with joint swelling, forehead furrowing, and significant clubbing in all fingers and toes. Currently, no standard treatments are approved for this disease; medical care is palliative and includes non-steroidal anti-inflammatory drugs, corticosteroids, tamoxifen, retinoids, and risedronate. Colchicine may be helpful for the pain due to subperiosteal new bone formation. Our patient was treated with etoricoxib 60 mg once daily and showed a significant clinical improvement at the 6-month mark that was reversed upon the withdrawal of this medication. This case report highlights the importance of placing etoricoxib among first-line therapy recommendations for cases with confirmed primary hypertrophic osteoarthropathy diagnosis. To the best of our knowledge, this is the only case of primary hypertrophic osteoarthropathy from the Middle Eastern population of Arab ethnicity that has responded to non-steroidal anti-inflammatory drug therapy.

SELECTION OF CITATIONS
SEARCH DETAIL