Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 52
Filter
Add more filters

Country/Region as subject
Publication year range
1.
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
2.
Am J Med Genet A ; 194(1): 59-63, 2024 Jan.
Article in English | MEDLINE | ID: mdl-37698259

ABSTRACT

Childhood-onset cardiomyopathy is a genetically heterogeneous group of conditions with several genes implicated. Recently, biallelic loss-of-function variants in PPP1R13L have been reported in association with a syndromic form of dilated cardiomyopathy (DCM). In addition, affected children manifest skin and hair abnormalities, cleft lip and palate (CLP), and eye findings. Here, we delineate the condition further by describing the phenotype associated with a homozygous frameshift variant (p.Arg330 ProfsTer76) in PPP1R13L detected in two sibships in a consanguineous family with six affected children. The index case had DCM and wooly hair, two of his siblings had DCM and CLP while three cousins had, in addition, glaucoma. Global developmental delay was observed in one child. All the children, except one, died during early childhood. Whole exome sequencing and whole genome sequencing did not reveal any other plausible variant. We provide further evidence that implicates PPP1R13L in a variable syndromic form of severe childhood-onset DCM and suggests expanding the spectrum of this condition to include glaucoma. Given the variability of the phenotype associated with PPP1R13-related DCM, a thorough evaluation of each case is highly recommended even in the presence of an apparently isolated DCM.


Subject(s)
Cardiomyopathy, Dilated , Cleft Lip , Cleft Palate , Glaucoma , Child , Humans , Child, Preschool , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/genetics , Alleles , Cleft Lip/genetics , Cleft Palate/genetics , Phenotype , Glaucoma/genetics , Repressor Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics
3.
Pediatr Cardiol ; 43(6): 1373-1378, 2022 Aug.
Article in English | MEDLINE | ID: mdl-35257196

ABSTRACT

The aim of this study is to examine the possible high association between multiple ventricular septal defect (mVSDs) and noncompaction cardiomyopathy (NCM) as same embryological origin, and the effect of depressed ventricular function in NCM cases during the follow-up, using echocardiography. A total of 150 patients with mVSDs were diagnosed in a single center in Saudi Arabia; 40 cases with isolated or associated with minor congenital heart disease were recruited. Three specialist echocardiography consultants confirmed the NCM diagnosis separately using Jenni, Chin and Patrick criteria, and myocardial function was estimated by ejection fraction at admission and at follow-up after surgery. Stata-14 to analyze the data was used. In our cohort of 40 cases with mVSD (median age at diagnosis = 0.5 years; mean follow-up = 4.84 years), 13(33%) had criteria of non-compaction confirmed by the three specialist consultants. All were operated by surgery and 11 hybrid approach (interventional & surgery). A significant relationship between abnormal trabeculations and mVSD with or without non-compaction was observed, 34% vs 66% respectively (p < 0.03, Fisher's exact test). A repeated-measures t-test found the difference between follow-up and preoperative ejection-fractions to be statistically significant (t (39) = 2.07, p < 0.04). Further, the myocardial function in the mVSD non-compaction group normalized substantially postoperatively compared with preoperative assessment (mean difference (MD) 11.77, 95% CI: 4.40-19.14), whilst the mVSD group with normal myocardium had no significant change in the myocardium function (MD 0.74, 95% CI: -4.10-5.58). Thus, treatment outcome appears better in the mVSD non-compaction group than their peers with normal myocardium. Acknowledging the lack of genetic data, it is evident the high incidence of non-compaction in this cohort of patients with mVSD and supports our hypothesis of embryonic/genetic link, unlikely to be explained by acquired cardiomyopathy.


Subject(s)
Heart Defects, Congenital , Heart Septal Defects, Ventricular , Isolated Noncompaction of the Ventricular Myocardium , Echocardiography , Heart Defects, Congenital/diagnostic imaging , Heart Septal Defects, Ventricular/diagnostic imaging , Heart Septal Defects, Ventricular/surgery , Humans , Isolated Noncompaction of the Ventricular Myocardium/diagnostic imaging , Myocardium , Ventricular Function, Left
4.
Genet Med ; 23(8): 1551-1568, 2021 08.
Article in English | MEDLINE | ID: mdl-33875846

ABSTRACT

PURPOSE: Within this study, we aimed to discover novel gene-disease associations in patients with no genetic diagnosis after exome/genome sequencing (ES/GS). METHODS: We followed two approaches: (1) a patient-centered approach, which after routine diagnostic analysis systematically interrogates variants in genes not yet associated to human diseases; and (2) a gene variant centered approach. For the latter, we focused on de novo variants in patients that presented with neurodevelopmental delay (NDD) and/or intellectual disability (ID), which are the most common reasons for genetic testing referrals. Gene-disease association was assessed using our data repository that combines ES/GS data and Human Phenotype Ontology terms from over 33,000 patients. RESULTS: We propose six novel gene-disease associations based on 38 patients with variants in the BLOC1S1, IPO8, MMP15, PLK1, RAP1GDS1, and ZNF699 genes. Furthermore, our results support causality of 31 additional candidate genes that had little published evidence and no registered OMIM phenotype (56 patients). The phenotypes included syndromic/nonsyndromic NDD/ID, oral-facial-digital syndrome, cardiomyopathies, malformation syndrome, short stature, skeletal dysplasia, and ciliary dyskinesia. CONCLUSION: Our results demonstrate the value of data repositories which combine clinical and genetic data for discovering and confirming gene-disease associations. Genetic laboratories should be encouraged to pursue such analyses for the benefit of undiagnosed patients and their families.


Subject(s)
Exome , Intellectual Disability , Base Sequence , Exome/genetics , Humans , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Nerve Tissue Proteins , Phenotype , Exome Sequencing
5.
Pacing Clin Electrophysiol ; 44(2): 380-384, 2021 02.
Article in English | MEDLINE | ID: mdl-32965045

ABSTRACT

Sick sinus syndrome (SSS) is a group of disorders characterized by an abnormal cardiac impulse formation or propagation from the sinoatrial node. Mutated SCN5A has been reported in SSS, however, homozygosity of SCN5A is exceedingly rare. Here, we report a consanguineous family with four affected children with SSS. Symptomatic bradycardia necessitated implanting a pacemaker in all of them. Sequencing SCN5A revealed a novel homozygous variant (p.Cys1850Arg), which was predicted to interfere with protein folding. Our report describes the phenotype of a novel homozygous SCN5A variant and contributes to the compendium of molecular pathology of inherited arrhythmias in consanguineous populations.


Subject(s)
Mutation , NAV1.5 Voltage-Gated Sodium Channel/genetics , Sick Sinus Syndrome/genetics , Adolescent , Female , Homozygote , Humans , Infant , Male , Pacemaker, Artificial , Pedigree , Sick Sinus Syndrome/therapy , Young Adult
6.
Monaldi Arch Chest Dis ; 92(3)2021 Dec 29.
Article in English | MEDLINE | ID: mdl-34964577

ABSTRACT

Hypertrophic cardiomyopathy (HCM) is a group of heterogeneous disorders that are most commonly passed on in a heritable manner. It is a relatively rare disease around the globe, but due to increased rates of consanguinity within the Kingdom of Saudi Arabia, we speculate a high incidence of undiagnosed cases. The aim of this paper is to elucidate a systematic approach in dealing with HCM patients and since HCM has variable presentation, we have summarized differentials for diagnosis and how different subtypes and genes can have an impact on the clinical picture, management and prognosis. Moreover, we propose a referral multi-disciplinary team HCM-Family Unit in Saudi Arabia and an integrated role in a network between King Faisal Hospital and Inherited and Rare Cardiovascular Disease Unit-Monaldi Hospital, Italy (among the 24 excellence centers of the European Reference Network (ERN) GUARD-Heart).   Graphical Abstract.


Subject(s)
Cardiomyopathy, Hypertrophic , Cardiomyopathy, Hypertrophic/diagnosis , Cardiomyopathy, Hypertrophic/epidemiology , Cardiomyopathy, Hypertrophic/genetics , Humans , Italy/epidemiology , Prognosis
7.
Eur Heart J ; 40(35): 2964-2975, 2019 09 14.
Article in English | MEDLINE | ID: mdl-31170290

ABSTRACT

AIMS: Calmodulinopathies are rare life-threatening arrhythmia syndromes which affect mostly young individuals and are, caused by mutations in any of the three genes (CALM 1-3) that encode identical calmodulin proteins. We established the International Calmodulinopathy Registry (ICalmR) to understand the natural history, clinical features, and response to therapy of patients with a CALM-mediated arrhythmia syndrome. METHODS AND RESULTS: A dedicated Case Report File was created to collect demographic, clinical, and genetic information. ICalmR has enrolled 74 subjects, with a variant in the CALM1 (n = 36), CALM2 (n = 23), or CALM3 (n = 15) genes. Sixty-four (86.5%) were symptomatic and the 10-year cumulative mortality was 27%. The two prevalent phenotypes are long QT syndrome (LQTS; CALM-LQTS, n = 36, 49%) and catecholaminergic polymorphic ventricular tachycardia (CPVT; CALM-CPVT, n = 21, 28%). CALM-LQTS patients have extremely prolonged QTc intervals (594 ± 73 ms), high prevalence (78%) of life-threatening arrhythmias with median age at onset of 1.5 years [interquartile range (IQR) 0.1-5.5 years] and poor response to therapies. Most electrocardiograms (ECGs) show late onset peaked T waves. All CALM-CPVT patients were symptomatic with median age of onset of 6.0 years (IQR 3.0-8.5 years). Basal ECG frequently shows prominent U waves. Other CALM-related phenotypes are idiopathic ventricular fibrillation (IVF, n = 7), sudden unexplained death (SUD, n = 4), overlapping features of CPVT/LQTS (n = 3), and predominant neurological phenotype (n = 1). Cardiac structural abnormalities and neurological features were present in 18 and 13 patients, respectively. CONCLUSION: Calmodulinopathies are largely characterized by adrenergically-induced life-threatening arrhythmias. Available therapies are disquietingly insufficient, especially in CALM-LQTS. Combination therapy with drugs, sympathectomy, and devices should be considered.


Subject(s)
Arrhythmias, Cardiac/genetics , DNA Mutational Analysis , Genetic Variation/genetics , Registries , Age of Onset , Arrhythmias, Cardiac/mortality , Calmodulin/genetics , Child , Child, Preschool , Death, Sudden, Cardiac/etiology , Female , Humans , Long QT Syndrome/genetics , Phenotype , Survival Rate , Tachycardia, Ventricular/genetics
8.
Genet Med ; 21(1): 185-188, 2019 01.
Article in English | MEDLINE | ID: mdl-29892088

ABSTRACT

PURPOSE: Wolf-Hirschhorn syndrome (WHS) is a genomic disorder with a recognizable dysmorphology profile caused by hemizygosity at 4p16.3. Previous attempts have failed to map the minimal critical locus to a single gene, leaving open the possibility that the core phenotypic components of the syndrome are caused by the combined haploinsufficiency of multiple genes. METHODS: Clinical exome sequencing and "reverse" phenotyping. RESULTS: We identified two patients with de novo truncating variants in WHSC1, which maps to the WHS critical locus. The phenotype of these two individuals is consistent with WHS, which suggests that haploinsufficiency of WHSC1 is sufficient to recapitulate the core phenotype (characteristic facies, and growth and developmental delay) of this classic microdeletion syndrome. CONCLUSION: Our study expands the list of microdeletion syndromes that are solved at the single-gene level, and establishes WHSC1 as a disease gene in humans. Given the severe nature of the reported variants, the full phenotypic expression of WHSC1 may be further expanded by future reports of milder variants.


Subject(s)
Developmental Disabilities/genetics , Genetic Predisposition to Disease , Histone-Lysine N-Methyltransferase/genetics , Repressor Proteins/genetics , Wolf-Hirschhorn Syndrome/genetics , Child, Preschool , Chromosome Deletion , Comparative Genomic Hybridization , Developmental Disabilities/diagnosis , Developmental Disabilities/pathology , Female , Haploinsufficiency/genetics , Humans , Male , Phenotype , Wolf-Hirschhorn Syndrome/diagnosis , Wolf-Hirschhorn Syndrome/pathology
9.
BMC Med Genet ; 20(1): 12, 2019 01 11.
Article in English | MEDLINE | ID: mdl-30635043

ABSTRACT

BACKGROUND: Rubinstein-Taybi syndrome (RSTS) Type 1 (OMIM 180849) is characterized by three main features: intellectual disability; broad and frequently angulated thumbs and halluces; and characteristic facial dysmorphism. CASE PRESENTATION: We report on a Saudi boy with RSTS Type 1 and the following distinct features: a midline notch of the upper lip, a bifid tip of the tongue, a midline groove of the lower lip, plump fingers with broad / flat fingertips, and brachydactyly. The child was found to be heterozygous in the CREBBP gene for a sequence variant designated c.4963del, which is predicted to result in premature protein termination p.Leu1655Cysfs*89. The child and his father were also found to be heterozygous in the EP300 gene for a sequence variant designated c.586A > G, which is predicted to result in the amino-acid substitution p.Ile196Val. CONCLUSION: Our report expands the clinical spectrum of RSTS to include several distinct facial and limb features. The variant of the CREBBP gene is known to be causative of RSTS Type 1. The variant in the EP300 gene is benign since the father carried the same variant and exhibited no abnormalities. However, functional studies are required to investigate if this benign EP300 variant influences the phenotype in the presence of disease-causing CREBBP gene mutations.


Subject(s)
CREB-Binding Protein/genetics , E1A-Associated p300 Protein/genetics , Genetic Association Studies , Rubinstein-Taybi Syndrome/genetics , Child, Preschool , Exons , Genetic Testing , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Male , Mutation , Phenotype , Rubinstein-Taybi Syndrome/physiopathology , Saudi Arabia , Sequence Analysis, DNA
10.
Clin Genet ; 95(2): 310-319, 2019 02.
Article in English | MEDLINE | ID: mdl-30561787

ABSTRACT

Defects in the peroxisomes biogenesis and/or function result in peroxisomal disorders. In this study, we describe the largest Arab cohort to date (72 families) of clinically, biochemically and molecularly characterized patients with peroxisomal disorders. At the molecular level, we identified 43 disease-causing variants, half of which are novel. The founder nature of many of the variants allowed us to calculate the minimum disease burden for these disorders in our population ~1:30 000, which is much higher than previous estimates in other populations. Clinically, we found an interesting trend toward genotype/phenotype correlation in terms of long-term survival. Nearly half (40/75) of our peroxisomal disorders patients had documented survival beyond 1 year of age. Most unusual among the long-term survivors was a multiplex family in which the affected members presented as adults with non-specific intellectual disability and epilepsy. Other unusual presentations included the very recently described peroxisomal fatty acyl-CoA reductase 1 disorder as well as CRD, spastic paraparesis, white matter (CRSPW) syndrome. We conclude that peroxisomal disorders are highly heterogeneous in their clinical presentation. Our data also confirm the demonstration that milder forms of Zellweger spectrum disorders cannot be ruled out by the "gold standard" very long chain fatty acids assay, which highlights the value of a genomics-first approach in these cases.


Subject(s)
Arabs , Peroxisomal Disorders/epidemiology , Peroxisomal Disorders/etiology , Arabs/genetics , Biomarkers , Brain/abnormalities , Brain/diagnostic imaging , Cohort Studies , Consanguinity , Cost of Illness , Disease Management , Disease Susceptibility , Facies , Female , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging , Male , Mutation , Pedigree , Peroxisomal Disorders/diagnosis , Peroxisomal Disorders/therapy , Phenotype , Population Surveillance , Prognosis
11.
Am J Med Genet A ; 176(3): 715-721, 2018 03.
Article in English | MEDLINE | ID: mdl-29383837

ABSTRACT

Temtamy syndrome is a syndromic form of intellectual disability characterized by ocular involvement, epilepsy and dysgenesis of the corpus callosum. After we initially mapped the disease to C12orf57, we noted a high carrier frequency of an ancient startloss founder mutation [c.1A>G; p.M1?] in our population, and variable phenotypic expressivity in newly identified cases. This study aims to combine 33 previously published patients with 23 who are described here for the first time to further delineate the phenotype of this syndrome. In addition to the known p.M1? founder, we describe four novel homozygous variants, thus increasing the number of Temtamy syndrome-related C12orf57 variants to seven, all but one predicted to be loss of function. While all patients presented with intellectual disability/developmental delay, the frequency of other phenotypic features was variable: 73.2% (41/56) had epilepsy, 63% (34/54) had corpus callosal abnormalities, 14.5% (8/55) had coloboma, and 16.4% (9/55) had microphthalmia. Our analysis also revealed a high frequency of less recognized features such as congenital heart disease (51.4%), and brain white matter abnormalities (38%, 19/50). We conclude that C12orf57 variants should be considered in the etiology of developmental delay/intellectual disability, even when typical syndromic features are lacking, especially in those who trace their ancestry to Saudi Arabia where a founder C12orf57 mutation is among the most common recessive causes of intellectual disability.


Subject(s)
Agenesis of Corpus Callosum/diagnosis , Coloboma/diagnosis , Craniofacial Abnormalities/diagnosis , Eye Abnormalities/diagnosis , Agenesis of Corpus Callosum/epidemiology , Agenesis of Corpus Callosum/genetics , Alleles , Coloboma/epidemiology , Coloboma/genetics , Craniofacial Abnormalities/epidemiology , Craniofacial Abnormalities/genetics , Eye Abnormalities/genetics , Facies , Genetic Association Studies , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging , Mutation , Phenotype , Prevalence
12.
Hum Mutat ; 38(12): 1649-1659, 2017 12.
Article in English | MEDLINE | ID: mdl-28940506

ABSTRACT

F-box and leucine-rich repeat protein 4 (FBXL4) is a mitochondrial protein whose exact function is not yet known. However, cellular studies have suggested that it plays significant roles in mitochondrial bioenergetics, mitochondrial DNA (mtDNA) maintenance, and mitochondrial dynamics. Biallelic pathogenic variants in FBXL4 are associated with an encephalopathic mtDNA maintenance defect syndrome that is a multisystem disease characterized by lactic acidemia, developmental delay, and hypotonia. Other features are feeding difficulties, growth failure, microcephaly, hyperammonemia, seizures, hypertrophic cardiomyopathy, elevated liver transaminases, recurrent infections, variable distinctive facial features, white matter abnormalities and cerebral atrophy found in neuroimaging, combined deficiencies of multiple electron transport complexes, and mtDNA depletion. Since its initial description in 2013, 36 different pathogenic variants in FBXL4 were reported in 50 affected individuals. In this report, we present 37 additional affected individuals and 11 previously unreported pathogenic variants. We summarize the clinical features of all 87 individuals with FBXL4-related mtDNA maintenance defect, review FBXL4 structure and function, map the 47 pathogenic variants onto the gene structure to assess the variants distribution, and investigate the genotype-phenotype correlation. Finally, we provide future directions to understand the disease mechanism and identify treatment strategies.


Subject(s)
DNA, Mitochondrial/genetics , F-Box Proteins/genetics , Genetic Association Studies , Mitochondrial Encephalomyopathies/genetics , Ubiquitin-Protein Ligases/genetics , Acidosis, Lactic/genetics , Cardiomyopathy, Hypertrophic/genetics , Genetic Predisposition to Disease , Humans , Kaplan-Meier Estimate , Mitochondria/genetics , Mitochondrial Encephalomyopathies/epidemiology , Mitochondrial Encephalomyopathies/pathology , Mitochondrial Proteins/genetics , Muscle Hypotonia/genetics , Mutation , Oxidative Phosphorylation , Proteome/genetics
13.
Genome Res ; 24(2): 291-9, 2014 Feb.
Article in English | MEDLINE | ID: mdl-24389050

ABSTRACT

Primordial dwarfism (PD) is a disease in which severely impaired fetal growth persists throughout postnatal development and results in stunted adult size. The condition is highly heterogeneous clinically, but the use of certain phenotypic aspects such as head circumference and facial appearance has proven helpful in defining clinical subgroups. In this study, we present the results of clinical and genomic characterization of 16 new patients in whom a broad definition of PD was used (e.g., 3M syndrome was included). We report a novel PD syndrome with distinct facies in two unrelated patients, each with a different homozygous truncating mutation in CRIPT. Our analysis also reveals, in addition to mutations in known PD disease genes, the first instance of biallelic truncating BRCA2 mutation causing PD with normal bone marrow analysis. In addition, we have identified a novel locus for Seckel syndrome based on a consanguineous multiplex family and identified a homozygous truncating mutation in DNA2 as the likely cause. An additional novel PD disease candidate gene XRCC4 was identified by autozygome/exome analysis, and the knockout mouse phenotype is highly compatible with PD. Thus, we add a number of novel genes to the growing list of PD-linked genes, including one which we show to be linked to a novel PD syndrome with a distinct facial appearance. PD is extremely heterogeneous genetically and clinically, and genomic tools are often required to reach a molecular diagnosis.


Subject(s)
Dwarfism/genetics , Microcephaly/genetics , Muscle Hypotonia/genetics , Mutation/genetics , Spine/abnormalities , Adaptor Proteins, Signal Transducing/genetics , BRCA2 Protein/genetics , Bone Marrow/pathology , DNA-Binding Proteins/genetics , Dwarfism/pathology , Facies , Female , Genetic Predisposition to Disease , Homozygote , Humans , Male , Microcephaly/pathology , Muscle Hypotonia/pathology , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Spine/pathology
14.
Cardiology ; 137(3): 188-192, 2017.
Article in English | MEDLINE | ID: mdl-28441660

ABSTRACT

OBJECTIVE: Cardiomyopathy (CMP) in children is a clinically and genetically heterogeneous group of disorders. Disease-associated mutations have been identified in more than 50 genes. Recently, mutations in the mitochondrial tRNA processing gene, ELAC2, were reported to be associated with the recessively inherited form of hypertrophic CMP (HCM). This study is aimed at describing the cardiac phenotype and outcome of ELAC2 mutation. METHODS: We performed whole exome sequencing followed by targeted mutation screening to identify the genetic etiology of severe infantile-onset CMP in 64 consanguineous Saudi families. RESULTS: A previously reported mutation (p.Phe154Leu) in ELAC2 gene was detected in 16 families. The index cases presented between 2 and 7 months of age with HCM in 13 infants and dilated CMP (DCM) in 3. Pericardial effusion was observed in 7 infants (44%). All infants died with a median age of death of 4 months. Almost 1/3 of them died during the initial presentation. CONCLUSION: Our study suggests screening the ELAC2 gene in severe infantile-onset HCM or DCM of unknown etiology, especially in the presence of pericardial effusion. Our work demonstrates a universally poor outcome of the (p.Phe154Leu) variant in ELAC2 gene; a correlation that helps in counseling parents and in planning appropriate medical intervention.


Subject(s)
Cardiomyopathy, Dilated/genetics , Cardiomyopathy, Hypertrophic/genetics , Neoplasm Proteins/genetics , Family Health , Female , Homozygote , Humans , Infant , Male , Mutation, Missense , Phenotype , Saudi Arabia
15.
Genome Res ; 23(2): 236-47, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23105016

ABSTRACT

Retinal dystrophy (RD) is a heterogeneous group of hereditary diseases caused by loss of photoreceptor function and contributes significantly to the etiology of blindness globally but especially in the industrialized world. The extreme locus and allelic heterogeneity of these disorders poses a major diagnostic challenge and often impedes the ability to provide a molecular diagnosis that can inform counseling and gene-specific treatment strategies. In a large cohort of nearly 150 RD families, we used genomic approaches in the form of autozygome-guided mutation analysis and exome sequencing to identify the likely causative genetic lesion in the majority of cases. Additionally, our study revealed six novel candidate disease genes (C21orf2, EMC1, KIAA1549, GPR125, ACBD5, and DTHD1), two of which (ACBD5 and DTHD1) were observed in the context of syndromic forms of RD that are described for the first time.


Subject(s)
Exome , Mutation , Retinal Dystrophies/genetics , Family , Genetic Association Studies , Genotype , Humans , Phenotype , Sequence Analysis, DNA
16.
BMC Med Genet ; 17: 3, 2016 Jan 14.
Article in English | MEDLINE | ID: mdl-26768247

ABSTRACT

BACKGROUND: Familial dilated cardiomyopathy (DCM) is genetically heterogeneous. Mutations in more than 40 genes have been identified in familial cases, mostly inherited in an autosomal dominant pattern. DCM due to recessive mutations is rarely observed. In consanguineous families, homozygosity mapping and whole exome sequencing (WES) can be utilized to identify the genetic defects in recessively inherited DCM. METHODS: In a consanguineous family with four affected siblings with severe DCM, we combined homozygosity mapping, linkage analysis and WES, to uncover the genetic defect. RESULTS: A region of homozygosity (ROH) on chromosome 8q24.13-24.23 was found to be shared by all of the four affected siblings. WES detected ~47,000 variants that were filtered to a homozygous mutation (p.Gly243Arg) in the FBXO32 gene, located within the identified ROH. The mutation segregated with the phenotype, replaced a highly-conserved amino acid, and was not detected in 1986 ethnically-matched chromosomes. FBXO32, which encodes a muscle-specific ubiquitin ligase, has been implicated in the pathogenesis of cardiomyopathy through the ubiquitin proteasome system (UPS). In addition, FBXO32-knockout mice manifest with cardiomyopathy. Screening the index patient for all of the WES variants in 48 genes known to be implicated in hypertrophic and dilated cardiomyopathy was negative. CONCLUSIONS: Our data suggest that FBXO32 is a candidate gene for recessive DCM. Acting as a cardiac ubiquitin ligase, mutated FBXO32 could perturb the degradation of target proteins in the UPS, the impairment of which has been observed in cardiomyopathy. Our work proposes that genes encoding other ubiquitin ligases could also be implicated in familial cardiomyopathy.


Subject(s)
Cardiomyopathy, Dilated/genetics , Heart , Muscle Proteins/genetics , SKP Cullin F-Box Protein Ligases/genetics , Adolescent , Adult , Amino Acid Sequence , Animals , Blood Pressure , Chromosome Mapping , Computational Biology , Exome , Female , Genetic Association Studies , Genetic Heterogeneity , Genetic Linkage , Homozygote , Humans , Mice, Knockout , Molecular Sequence Data , Mutation, Missense , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Protein Conformation , Young Adult
17.
J Med Genet ; 52(3): 186-94, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25539947

ABSTRACT

BACKGROUND: There are numerous nuclear genes that cause mitochondrial disorders and clinically and genetically heterogeneous disorders whose aetiology often remains unsolved. In this study, we aim to investigate an autosomal recessive syndrome causing leukodystrophy and neuroregression. We studied six patients from five unrelated consanguineous families. METHODS: Patients underwent full neurological, radiological, genetic, metabolic and dysmorphological examinations. Exome sequencing coupled with autozygosity mapping, Sanger sequencing, microsatellite haplotyping, standard and molecular karyotyping and whole mitochondrial DNA sequencing were used to identify the genetic cause of the syndrome. Immunohistochemistry, transmission electron microscopy, confocal microscopy, dipstick assays, quantitative PCR, reverse transcription PCR and quantitative reverse transcription PCR were performed on different tissue samples from the patients. RESULTS: We identified a homoallelic missense founder mutation in ISCA2 leading to mitochondrial depletion and reduced complex I activity as well as decreased ISCA2, ISCA1 and IBA57 expression in fibroblasts. MRI indicated similar white matter abnormalities in the patients. Histological examination of the skeletal muscle showed mild to moderate variation in myofibre size and the presence of many randomly distributed atrophic fibres. CONCLUSIONS: Our data demonstrate that ISCA2 deficiency leads to a hereditary mitochondrial neurodegenerative white matter disease in infancy.


Subject(s)
Alexander Disease/genetics , Iron-Sulfur Proteins/genetics , Mitochondrial Diseases/genetics , Neurodegenerative Diseases/genetics , Adult , Alexander Disease/physiopathology , Child, Preschool , DNA, Mitochondrial/genetics , Exome/genetics , Female , Humans , Infant , Male , Middle Aged , Mitochondrial Diseases/physiopathology , Mutation, Missense , Neurodegenerative Diseases/physiopathology , Pedigree , Sequence Analysis, DNA , White Matter/abnormalities , White Matter/metabolism
18.
Pacing Clin Electrophysiol ; 36(5): e140-2, 2013 May.
Article in English | MEDLINE | ID: mdl-22650415

ABSTRACT

Catecholaminergic polymorphic ventricular tachycardia (CPVT) manifests with episodic syncope or sudden death in young patients following physical activity or emotional stress. The autosomal recessive form of CPVT is caused by mutations in the CASQ2 gene. In a consanguineous family, a novel homozygous CASQ2 mutation (p.L77P) was identified in a child with CPVT who required implantation of a cardioverter defibrillator due to episodes of syncope while on medical therapy. Genetic testing found the younger sibling, who had normal initial clinical screening, to be affected. Our cases underscore the importance of family screening through genetic testing to preemptively apply the appropriate medical intervention in CPVT.


Subject(s)
Calsequestrin/genetics , Genetic Predisposition to Disease/genetics , Genetic Testing , Mutation/genetics , Polymorphism, Single Nucleotide/genetics , Tachycardia, Ventricular/diagnosis , Tachycardia, Ventricular/genetics , Adolescent , Genetic Carrier Screening , Heterozygote , Humans , Male , Phenotype , Saudi Arabia
19.
J Saudi Heart Assoc ; 35(2): 144-147, 2023.
Article in English | MEDLINE | ID: mdl-37325369

ABSTRACT

Primary carnitine deficiency (PCD) is an autosomal recessive disorder characterized by decreased carnitine levels essential for Beta oxidation in various organs, including the heart. Early diagnosis and treatment of PCD can revert cardiomyopathy. A 13-year-old girl presented with heart failure due to dilated cardiomyopathy and severe cardiac dysfunction; following L carnitine treatment, the patient's clinical conditions improved, and cardiac functions returned to normal within weeks. Investigations revealed PCD; regular L carnitine has been provided, all cardiac medications are discontinued, and the patient is doing well. We believe PCD should be ruled out in every patient with cardiomyopathy.

20.
JCI Insight ; 8(17)2023 09 08.
Article in English | MEDLINE | ID: mdl-37561591

ABSTRACT

Pediatric cardiomyopathy (CM) represents a group of rare, severe disorders that affect the myocardium. To date, the etiology and mechanisms underlying pediatric CM are incompletely understood, hampering accurate diagnosis and individualized therapy development. Here, we identified biallelic variants in the highly conserved flightless-I (FLII) gene in 3 families with idiopathic, early-onset dilated CM. We demonstrated that patient-specific FLII variants, when brought into the zebrafish genome using CRISPR/Cas9 genome editing, resulted in the manifestation of key aspects of morphological and functional abnormalities of the heart, as observed in our patients. Importantly, using these genetic animal models, complemented with in-depth loss-of-function studies, we provided insights into the function of Flii during ventricular chamber morphogenesis in vivo, including myofibril organization and cardiomyocyte cell adhesion, as well as trabeculation. In addition, we identified Flii function to be important for the regulation of Notch and Hippo signaling, crucial pathways associated with cardiac morphogenesis and function. Taken together, our data provide experimental evidence for a role for FLII in the pathogenesis of pediatric CM and report biallelic variants as a genetic cause of pediatric CM.


Subject(s)
Cardiomyopathies , Microfilament Proteins , Animals , Cell Adhesion/genetics , Microfilament Proteins/genetics , Myocytes, Cardiac/metabolism , Myofibrils/metabolism , Zebrafish/genetics , Trans-Activators , Cardiomyopathies/genetics
SELECTION OF CITATIONS
SEARCH DETAIL