Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 110(11): 1919-1937, 2023 11 02.
Article in English | MEDLINE | ID: mdl-37827158

ABSTRACT

Misregulation of histone lysine methylation is associated with several human cancers and with human developmental disorders. DOT1L is an evolutionarily conserved gene encoding a lysine methyltransferase (KMT) that methylates histone 3 lysine-79 (H3K79) and was not previously associated with a Mendelian disease in OMIM. We have identified nine unrelated individuals with seven different de novo heterozygous missense variants in DOT1L through the Undiagnosed Disease Network (UDN), the SickKids Complex Care genomics project, and GeneMatcher. All probands had some degree of global developmental delay/intellectual disability, and most had one or more major congenital anomalies. To assess the pathogenicity of the DOT1L variants, functional studies were performed in Drosophila and human cells. The fruit fly DOT1L ortholog, grappa, is expressed in most cells including neurons in the central nervous system. The identified DOT1L variants behave as gain-of-function alleles in flies and lead to increased H3K79 methylation levels in flies and human cells. Our results show that human DOT1L and fly grappa are required for proper development and that de novo heterozygous variants in DOT1L are associated with a Mendelian disease.


Subject(s)
Congenital Abnormalities , Developmental Disabilities , Histone-Lysine N-Methyltransferase , Humans , Gain of Function Mutation , Histone-Lysine N-Methyltransferase/genetics , Histones/genetics , Histones/metabolism , Lysine , Methylation , Methyltransferases/genetics , Neoplasms/genetics , Drosophila/genetics , Drosophila Proteins/genetics , Developmental Disabilities/genetics , Congenital Abnormalities/genetics
2.
Hum Mol Genet ; 30(23): 2300-2314, 2021 11 16.
Article in English | MEDLINE | ID: mdl-34245260

ABSTRACT

Here, we report on six unrelated individuals, all presenting with early-onset global developmental delay, associated with impaired motor, speech and cognitive development, partly with developmental epileptic encephalopathy and physical dysmorphisms. All individuals carry heterozygous missense variants of KCND2, which encodes the voltage-gated potassium (Kv) channel α-subunit Kv4.2. The amino acid substitutions associated with the variants, p.(Glu323Lys) (E323K), p.(Pro403Ala) (P403A), p.(Val404Leu) (V404L) and p.(Val404Met) (V404M), affect sites known to be critical for channel gating. To unravel their likely pathogenicity, recombinant mutant channels were studied in the absence and presence of auxiliary ß-subunits under two-electrode voltage clamp in Xenopus oocytes. All channel mutants exhibited slowed and incomplete macroscopic inactivation, and the P403A variant in addition slowed activation. Co-expression of KChIP2 or DPP6 augmented the functional expression of both wild-type and mutant channels; however, the auxiliary ß-subunit-mediated gating modifications differed from wild type and among mutants. To simulate the putative setting in the affected individuals, heteromeric Kv4.2 channels (wild type + mutant) were studied as ternary complexes (containing both KChIP2 and DPP6). In the heteromeric ternary configuration, the E323K variant exhibited only marginal functional alterations compared to homomeric wild-type ternary, compatible with mild loss-of-function. By contrast, the P403A, V404L and V404M variants displayed strong gating impairment in the heteromeric ternary configuration, compatible with loss-of-function or gain-of-function. Our results support the etiological involvement of Kv4.2 channel gating impairment in early-onset monogenic global developmental delay. In addition, they suggest that gain-of-function mechanisms associated with a substitution of V404 increase epileptic seizure susceptibility.


Subject(s)
Developmental Disabilities/etiology , Developmental Disabilities/metabolism , Genetic Variation , Ion Channel Gating , Shal Potassium Channels/genetics , Shal Potassium Channels/metabolism , Alleles , Amino Acid Substitution , Biomarkers , Developmental Disabilities/diagnosis , Disease Susceptibility , Female , Humans , Infant , Infant, Newborn , Male , Mutation , Phenotype , Protein Subunits , Shal Potassium Channels/chemistry
3.
Am J Hum Genet ; 105(1): 213-220, 2019 07 03.
Article in English | MEDLINE | ID: mdl-31230721

ABSTRACT

De novo variants represent a significant cause of neurodevelopmental delay and intellectual disability. A genetic basis can be identified in only half of individuals who have neurodevelopmental disorders (NDDs); this indicates that additional causes need to be elucidated. We compared the frequency of de novo variants in patient-parent trios with (n = 2,030) versus without (n = 2,755) NDDs. We identified de novo variants in TAOK1 (thousand and one [TAO] amino acid kinase 1), which encodes the serine/threonine-protein kinase TAO1, in three individuals with NDDs but not in persons who did not have NDDs. Through further screening and the use of GeneMatcher, five additional individuals with NDDs were found to have de novo variants. All eight variants were absent from gnomAD (Genome Aggregation Database). The variant carriers shared a non-specific phenotype of developmental delay, and six individuals had additional muscular hypotonia. We established a fibroblast line of one mutation carrier, and we demonstrated that reduced mRNA levels of TAOK1 could be increased upon cycloheximide treatment. These results indicate nonsense-mediated mRNA decay. Further, there was neither detectable phosphorylated TAO1 kinase nor phosphorylated tau in these cells, and mitochondrial morphology was altered. Knockdown of the ortholog gene Tao1 (Tao, CG14217) in Drosophila resulted in delayed early development. The majority of the Tao1-knockdown flies did not survive beyond the third instar larval stage. When compared to control flies, Tao1 knockdown flies revealed changed morphology of the ventral nerve cord and the neuromuscular junctions as well as a decreased number of endings (boutons). Furthermore, mitochondria in mutant flies showed altered distribution and decreased size in axons of motor neurons. Thus, we provide compelling evidence that de novo variants in TAOK1 cause NDDs.


Subject(s)
Drosophila melanogaster/growth & development , Exome/genetics , Mutation , Neurodevelopmental Disorders/etiology , Protein Serine-Threonine Kinases/genetics , Animals , Child , Drosophila melanogaster/genetics , Drosophila melanogaster/metabolism , Female , Heterozygote , Humans , Male , Neurodevelopmental Disorders/pathology , Phenotype , Exome Sequencing
4.
Am J Hum Genet ; 104(3): 530-541, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827496

ABSTRACT

Acetylation of the lysine residues in histones and other DNA-binding proteins plays a major role in regulation of eukaryotic gene expression. This process is controlled by histone acetyltransferases (HATs/KATs) found in multiprotein complexes that are recruited to chromatin by the scaffolding subunit transformation/transcription domain-associated protein (TRRAP). TRRAP is evolutionarily conserved and is among the top five genes intolerant to missense variation. Through an international collaboration, 17 distinct de novo or apparently de novo variants were identified in TRRAP in 24 individuals. A strong genotype-phenotype correlation was observed with two distinct clinical spectra. The first is a complex, multi-systemic syndrome associated with various malformations of the brain, heart, kidneys, and genitourinary system and characterized by a wide range of intellectual functioning; a number of affected individuals have intellectual disability (ID) and markedly impaired basic life functions. Individuals with this phenotype had missense variants clustering around the c.3127G>A p.(Ala1043Thr) variant identified in five individuals. The second spectrum manifested with autism spectrum disorder (ASD) and/or ID and epilepsy. Facial dysmorphism was seen in both groups and included upslanted palpebral fissures, epicanthus, telecanthus, a wide nasal bridge and ridge, a broad and smooth philtrum, and a thin upper lip. RNA sequencing analysis of skin fibroblasts derived from affected individuals skin fibroblasts showed significant changes in the expression of several genes implicated in neuronal function and ion transport. Thus, we describe here the clinical spectrum associated with TRRAP pathogenic missense variants, and we suggest a genotype-phenotype correlation useful for clinical evaluation of the pathogenicity of the variants.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autistic Disorder/etiology , Intellectual Disability/etiology , Mutation, Missense , Nuclear Proteins/genetics , Adolescent , Adult , Amino Acid Sequence , Autistic Disorder/metabolism , Autistic Disorder/pathology , Child , Child, Preschool , Female , Genetic Association Studies , Humans , Infant , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Prognosis , Sequence Homology , Syndrome , Young Adult
5.
Am J Med Genet A ; 188(1): 272-282, 2022 01.
Article in English | MEDLINE | ID: mdl-34515416

ABSTRACT

By clinical whole exome sequencing, we identified 12 individuals with ages 3 to 37 years, including three individuals from the same family, with a consistent phenotype of intellectual disability (ID), macrocephaly, and overgrowth of adenoid tissue. All 12 individuals harbored a rare heterozygous variant in ZBTB7A which encodes the transcription factor Zinc finger and BTB-domain containing protein 7A, known to play a role in lympho- and hematopoiesis. ID was generally mild. Fetal hemoglobin (HbF) fraction was elevated 2.2%-11.2% (reference value <2% in individuals > 6 months) in four of the five individuals for whom results were available. Ten of twelve individuals had undergone surgery at least once for lymphoid hypertrophy limited to the pharynx. In the most severely affected individual (individual 1), airway obstruction resulted in 17 surgical procedures before the age of 13 years. Sleep apnea was present in 8 of 10 individuals. In the nine unrelated individuals, ZBTB7A variants were novel and de novo. The six frameshift/nonsense and four missense variants were spread throughout the gene. This is the first report of a cohort of individuals with this novel syndromic neurodevelopmental disorder.


Subject(s)
Intellectual Disability , Megalencephaly , Neurodevelopmental Disorders , Cell Line, Tumor , DNA-Binding Proteins/genetics , Fetal Hemoglobin , Humans , Intellectual Disability/genetics , Lymphoid Tissue , Megalencephaly/genetics , Neurodevelopmental Disorders/genetics , Transcription Factors/genetics
6.
Neuroepidemiology ; 56(4): 271-282, 2022.
Article in English | MEDLINE | ID: mdl-35576897

ABSTRACT

BACKGROUND: Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disease that affects motor neurons. In Europe, disease-causing genetic variants have been identified in 40-70% of familial ALS patients and approximately 5% of sporadic ALS patients. In Norway, the contribution of genetic variants to ALS has not yet been studied. In light of the potential development of personalized medicine, knowledge of the genetic causes of ALS in a population is becoming increasingly important. The present study provides clinical and genetic data on familial and sporadic ALS patients in a Norwegian population-based cohort. METHODS: Blood samples and clinical information from ALS patients were obtained at all 17 neurological departments throughout Norway during a 2-year period. Genetic analysis of the samples involved expansion analysis of C9orf72 and exome sequencing targeting 30 known ALS-linked genes. The variants were classified using genotype-phenotype correlations and bioinformatics tools. RESULTS: A total of 279 ALS patients were included in the study. Of these, 11.5% had one or several family members affected by ALS, whereas 88.5% had no known family history of ALS. A genetic cause of ALS was identified in 31 individuals (11.1%), among which 18 (58.1%) were familial and 13 (41.9%) were sporadic. The most common genetic cause was the C9orf72 expansion (6.8%), which was identified in 8 familial and 11 sporadic ALS patients. Pathogenic or likely pathogenic variants of SOD1 and TBK1 were identified in 10 familial and 2 sporadic cases. C9orf72 expansions dominated in patients from the Northern and Central regions, whereas SOD1 variants dominated in patients from the South-Eastern region. CONCLUSION: In the present study, we identified several pathogenic gene variants in both familial and sporadic ALS patients. Restricting genetic analysis to only familial cases would miss more than 40 percent of those with a disease-causing genetic variant, indicating the need for genetic analysis in sporadic cases as well.


Subject(s)
Amyotrophic Lateral Sclerosis , Neurodegenerative Diseases , Amyotrophic Lateral Sclerosis/epidemiology , Amyotrophic Lateral Sclerosis/genetics , C9orf72 Protein/genetics , Humans , Molecular Epidemiology , Superoxide Dismutase-1/genetics
7.
Tidsskr Nor Laegeforen ; 142(4)2022 03 01.
Article in English, Norwegian | MEDLINE | ID: mdl-35239266

ABSTRACT

BACKGROUND: VEXAS syndrome (Vacuoles, E1 enzyme, X-linked, Autoinflammatory, Somatic syndrome) first described in 2020, is caused by a limited repertoire of somatic mutations in UBA1, a gene involved in the initiation of ubiquitination. Ubiquitination, adding an ubiquitin protein to a substrate protein, can have various effects on the substrate. Disruption of UBA1 function results in diverse clinical manifestations, mimicking a variety of disorders. CASE PRESENTATION: A man in his sixties presented with fever, chest pain, fatigue, pulmonary infiltrates and elevated acute phase reactants. Initially he was thought to have extra-cranial giant cell arteritis. When he developed ear and nose chondritis, a revised diagnosis of relapsing polychondritis was made. Subsequently he developed macrocytic anaemia and thrombocytopenia. His condition remained resistant to medical therapy and he died eight years after disease onset. Analysis of stored DNA revealed a somatic mutation in UBA1 confirming the diagnosis of VEXAS syndrome. INTERPRETATION: VEXAS syndrome is a newly identified inflammatory disorder due to an acquired mutation in haematopoietic bone marrow cells in older men. The syndrome may be misdiagnosed as treatment-refractory relapsing polychondritis, polyarteritis nodosa, Sweet syndrome or giant cell arteritis. We describe the first individual with molecularly confirmed VEXAS syndrome in Norway.


Subject(s)
Giant Cell Arteritis , Myelodysplastic Syndromes , Pancytopenia , Polychondritis, Relapsing , Aged , Bone Marrow Failure Disorders , Humans , Inflammation , Male , Myelodysplastic Syndromes/diagnosis , Myelodysplastic Syndromes/genetics , Polychondritis, Relapsing/complications , Polychondritis, Relapsing/diagnosis , Polychondritis, Relapsing/genetics , Ubiquitin-Activating Enzymes/genetics
8.
Tidsskr Nor Laegeforen ; 142(1)2022 01 11.
Article in English, Norwegian | MEDLINE | ID: mdl-35026081

ABSTRACT

BACKGROUND: Arthrogryposis multiplex congenita (AMC) is a descriptive term that encompasses a group of congenital, aetiologically heterogeneous conditions characterised by multiple joint contractions. CASE PRESENTATION: As a teenager, the index patient was told she had AMC, as did one of her parents. Subsequently, she wondered how her condition might evolve over time, since her affected parent had become wheelchair- dependent. Her history and clinical findings led to genetic testing which identified a causative variant in the COL6A2 gene, revealing an underlying diagnosis of Bethlem myopathy. INTERPRETATION: Adults who have rare monogenic disorders may lack an aetiological diagnosis because of limited access to genetic laboratory testing in the past. Advances in genetic laboratory diagnostics during the last 10−15 years have made testing more widely available. As exemplified by this case, molecular genetic diagnosis may provide benefits such as information concerning prognosis and treatment options.


Subject(s)
Arthrogryposis , Contracture , Muscular Dystrophies , Adolescent , Adult , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Female , Genetic Testing , Humans , Muscle Weakness/diagnosis , Muscle Weakness/genetics
9.
J Hum Genet ; 66(11): 1101-1112, 2021 Nov.
Article in English | MEDLINE | ID: mdl-33980986

ABSTRACT

RBL2/p130, a member of the retinoblastoma family of proteins, is a key regulator of cell division and propagates irreversible senescence. RBL2/p130 is also involved in neuronal differentiation and survival, and eliminating Rbl2 in certain mouse strains leads to embryonic lethality accompanied by an abnormal central nervous system (CNS) phenotype. Conflicting reports exist regarding a role of RBL2/p130 in transcriptional regulation of DNA methyltransferases (DNMTs), as well as the control of telomere length. Here we describe the phenotype of three patients carrying bi-allelic RBL2-truncating variants. All presented with infantile hypotonia, severe developmental delay and microcephaly. Malignancies were not reported in carriers or patients. Previous studies carried out on mice and human cultured cells, associated RBL2 loss to DNA methylation and telomere length dysregulation. Here, we investigated whether patient cells lacking RBL2 display related abnormalities. The study of primary patient fibroblasts did not detect abnormalities in expression of DNMTs. Furthermore, methylation levels of whole genome DNA, and specifically of pericentromeric repeats and subtelomeric regions, were unperturbed. RBL2-null fibroblasts show no evidence for abnormal elongation by telomeric recombination. Finally, gradual telomere shortening, and normal onset of senescence were observed following continuous culturing of RBL2-mutated fibroblasts. Thus, this study resolves uncertainties regarding a potential non-redundant role for RBL2 in DNA methylation and telomere length regulation, and indicates that loss of function variants in RBL2 cause a severe autosomal recessive neurodevelopmental disorder in humans.


Subject(s)
Cognitive Dysfunction/genetics , DNA Methylation/genetics , Retinoblastoma-Like Protein p130/genetics , Telomere Shortening/genetics , Adolescent , Adult , Alleles , Animals , Child , Cognitive Dysfunction/complications , Cognitive Dysfunction/physiopathology , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Female , Fibroblasts/metabolism , Genetic Predisposition to Disease , Humans , Male , Methyltransferases/genetics , Mice , Microcephaly/complications , Microcephaly/genetics , Microcephaly/physiopathology , Motor Activity/physiology , Muscle Hypotonia/complications , Muscle Hypotonia/genetics , Muscle Hypotonia/physiopathology , Telomere/genetics , Exome Sequencing
10.
Am J Hum Genet ; 101(5): 768-788, 2017 Nov 02.
Article in English | MEDLINE | ID: mdl-29100089

ABSTRACT

Calcium/calmodulin-dependent protein kinase II (CAMK2) is one of the first proteins shown to be essential for normal learning and synaptic plasticity in mice, but its requirement for human brain development has not yet been established. Through a multi-center collaborative study based on a whole-exome sequencing approach, we identified 19 exceedingly rare de novo CAMK2A or CAMK2B variants in 24 unrelated individuals with intellectual disability. Variants were assessed for their effect on CAMK2 function and on neuronal migration. For both CAMK2A and CAMK2B, we identified mutations that decreased or increased CAMK2 auto-phosphorylation at Thr286/Thr287. We further found that all mutations affecting auto-phosphorylation also affected neuronal migration, highlighting the importance of tightly regulated CAMK2 auto-phosphorylation in neuronal function and neurodevelopment. Our data establish the importance of CAMK2A and CAMK2B and their auto-phosphorylation in human brain function and expand the phenotypic spectrum of the disorders caused by variants in key players of the glutamatergic signaling pathway.


Subject(s)
Calcium-Calmodulin-Dependent Protein Kinase Type 2/genetics , Intellectual Disability/genetics , Mutation/genetics , Animals , Brain/pathology , Cell Line , Exome/genetics , Female , Glutamic Acid/genetics , HEK293 Cells , Humans , Male , Mice , Mice, Inbred C57BL , Neurons/pathology , Phosphorylation/genetics , Signal Transduction/genetics
11.
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
12.
Genet Med ; 21(3): 663-675, 2019 03.
Article in English | MEDLINE | ID: mdl-30158690

ABSTRACT

PURPOSE: Defects in the cohesin pathway are associated with cohesinopathies, notably Cornelia de Lange syndrome (CdLS). We aimed to delineate pathogenic variants in known and candidate cohesinopathy genes from a clinical exome perspective. METHODS: We retrospectively studied patients referred for clinical exome sequencing (CES, N = 10,698). Patients with causative variants in novel or recently described cohesinopathy genes were enrolled for phenotypic characterization. RESULTS: Pathogenic or likely pathogenic single-nucleotide and insertion/deletion variants (SNVs/indels) were identified in established disease genes including NIPBL (N = 5), SMC1A (N = 14), SMC3 (N = 4), RAD21 (N = 2), and HDAC8 (N = 8). The phenotypes in this genetically defined cohort skew towards the mild end of CdLS spectrum as compared with phenotype-driven cohorts. Candidate or recently reported cohesinopathy genes were supported by de novo SNVs/indels in STAG1 (N = 3), STAG2 (N = 5), PDS5A (N = 1), and WAPL (N = 1), and one inherited SNV in PDS5A. We also identified copy-number deletions affecting STAG1 (two de novo, one of unknown inheritance) and STAG2 (one of unknown inheritance). Patients with STAG1 and STAG2 variants presented with overlapping features yet without characteristic facial features of CdLS. CONCLUSION: CES effectively identified disease-causing alleles at the mild end of the cohensinopathy spectrum and enabled characterization of candidate disease genes.


Subject(s)
Biological Variation, Population/genetics , Cell Cycle Proteins/genetics , Chromosomal Proteins, Non-Histone/genetics , Adolescent , Alleles , Antigens, Nuclear/genetics , Carrier Proteins/genetics , Child , Child, Preschool , Cohort Studies , De Lange Syndrome/diagnosis , De Lange Syndrome/genetics , Exome/genetics , Female , Gene Frequency/genetics , Genetic Heterogeneity , Humans , INDEL Mutation/genetics , Male , Mutation , Nuclear Proteins/genetics , Phenotype , Polymorphism, Single Nucleotide/genetics , Proto-Oncogene Proteins/genetics , Retrospective Studies , Exome Sequencing/methods , Cohesins
13.
Am J Med Genet A ; 179(9): 1884-1894, 2019 09.
Article in English | MEDLINE | ID: mdl-31313512

ABSTRACT

Brachyolmia is a skeletal dysplasia characterized by short spine-short stature, platyspondyly, and minor long bone abnormalities. We describe 18 patients, from different ethnic backgrounds and ages ranging from infancy to 19 years, with the autosomal recessive form, associated with PAPSS2. The main clinical features include disproportionate short stature with short spine associated with variable symptoms of pain, stiffness, and spinal deformity. Eight patients presented prenatally with short femora, whereas later in childhood their short-spine phenotype emerged. We observed the same pattern of changing skeletal proportion in other patients. The radiological findings included platyspondyly, irregular end plates of the elongated vertebral bodies, narrow disc spaces and short over-faced pedicles. In the limbs, there was mild shortening of femoral necks and tibiae in some patients, whereas others had minor epiphyseal or metaphyseal changes. In all patients, exome and Sanger sequencing identified homozygous or compound heterozygous PAPSS2 variants, including c.809G>A, common to white European patients. Bi-parental inheritance was established where possible. Low serum DHEAS, but not overt androgen excess was identified. Our study indicates that autosomal recessive brachyolmia occurs across continents and may be under-recognized in infancy. This condition should be considered in the differential diagnosis of short femora presenting in the second trimester.


Subject(s)
Dwarfism/genetics , Multienzyme Complexes/genetics , Musculoskeletal Abnormalities/genetics , Osteochondrodysplasias/genetics , Sulfate Adenylyltransferase/genetics , Adolescent , Adult , Child , Child, Preschool , Dwarfism/diagnostic imaging , Dwarfism/physiopathology , Female , Genes, Recessive/genetics , Genetic Predisposition to Disease , Homozygote , Humans , Infant , Infant, Newborn , Male , Musculoskeletal Abnormalities/diagnostic imaging , Musculoskeletal Abnormalities/physiopathology , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/physiopathology , Pedigree , Radiography , Spine/diagnostic imaging , Spine/physiopathology , Exome Sequencing , Young Adult
14.
Mol Genet Metab ; 121(1): 51-56, 2017 05.
Article in English | MEDLINE | ID: mdl-28377240

ABSTRACT

BACKGROUND: Rare sequence variants in at least five genes are known to cause monogenic obesity. In this study we aimed to investigate the prevalence of, and characterize, rare coding and splice site variants in LEP, LEPR, MC4R, PCSK1 and POMC in patients with morbid obesity and normal weight controls. METHOD: Targeted next-generation sequencing of all exons in LEP, LEPR, MC4R, PCSK1 and POMC was performed in 485 patients with morbid obesity and 327 normal weight population-based controls from Norway. RESULTS: In total 151 variants were detected. Twenty-eight (18.5%) of these were rare, coding or splice variants and five (3.3%) were novel. All individuals, except one control, were heterozygous for the 28 variants, and the distribution of the rare variants showed a significantly higher carrier frequency among cases than controls (9.9% vs. 4.9%, p=0.011). Four variants in MC4R were classified as pathogenic or likely pathogenic. CONCLUSION: Four cases (0.8%) of monogenic obesity were detected, all due to MC4R variants previously linked to monogenic obesity. Significant differences in carrier frequencies among patients with morbid obesity and normal weight controls suggest an association between heterozygous rare coding variants in these five genes and morbid obesity. However, additional studies in larger cohorts and functional testing of the novel variants identified are required to confirm the findings.


Subject(s)
Genetic Variation , High-Throughput Nucleotide Sequencing/methods , Obesity, Morbid/genetics , Sequence Analysis, DNA/methods , Adolescent , Adult , Age Distribution , Case-Control Studies , Child , Female , Genetic Predisposition to Disease , Humans , Leptin/genetics , Male , Middle Aged , Mutation Rate , Norway , Pro-Opiomelanocortin/genetics , Proprotein Convertase 1/genetics , Receptor, Melanocortin, Type 4/genetics , Receptors, Leptin/genetics , Young Adult
18.
Biochem J ; 457(1): 99-105, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24144304

ABSTRACT

PCSK9 (proprotein convertase subtilisin/kexin type 9) binds to the LDLR (low-density lipoprotein receptor) at the cell surface and disrupts recycling of the LDLR. However, PCSK9 also interacts with the LDLR in the ER (endoplasmic reticulum). In the present study we have investigated the role of PCSK9 for the transport of the LDLR from the ER to the cell membrane. A truncated LDLR consisting of the ectodomain (ED-LDLR) was used for these studies to avoid PCSK9-mediated degradation of the LDLR. The amount of secreted ED-LDLR was used as a measure of the amount of ED-LDLR transported from the ER. From co-transfection experiments of various PCSK9 and ED-LDLR plasmids, PCSK9 increased the amount of WT (wild-type) ED-LDLR in the medium, but not of an ED-LDLR lacking the EGF (epidermal growth factor)-A repeat or of a Class 2a mutant ED-LDLR which fails to exit the ER. Mutant PCSK9s which failed to undergo autocatalytic cleavage or failed to exit the ER, failed to increase the amount of WT-ED-LDLR in the medium. These mutants also reduced the amount of WT-ED-LDLR intracellularly, which could partly be prevented by the proteasome inhibitor lactacystine. WT-ED-LDLR promoted autocatalytic cleavage of pro-PCSK9. The findings of the present study indicate that the binding of WT-ED-LDLR to pro-PCSK9 in the ER promotes autocatalytic cleavage of PCSK9, and autocatalytically cleaved PCSK9 acts as a chaperone to promote the exit of WT-ED-LDLR from the ER.


Subject(s)
Endoplasmic Reticulum/metabolism , Molecular Chaperones/physiology , Proprotein Convertases/physiology , Receptors, LDL/metabolism , Serine Endopeptidases/physiology , Catalysis , Cell Membrane/metabolism , Hep G2 Cells , Homeostasis , Humans , Proprotein Convertase 9 , Proprotein Convertases/chemistry , Protein Interaction Domains and Motifs/physiology , Protein Isoforms/chemistry , Protein Isoforms/physiology , Protein Precursors/chemistry , Protein Precursors/physiology , Protein Transport , Proteolysis , Receptors, LDL/chemistry , Serine Endopeptidases/chemistry
19.
Tidsskr Nor Laegeforen ; 135(20): 1833-7, 2015 Nov 03.
Article in English, Norwegian | MEDLINE | ID: mdl-26534809

ABSTRACT

BACKGROUND: New DNA-sequencing technology is revolutionising medical diagnostics. Through the use of exome sequencing, it is now possible to sequence all human genes in parallel. This technology has been widely used in research over the last few years and is now also being applied to diagnostics. The aim of this study was to systematically examine initial experiences with diagnostic exome sequencing in Norway. MATERIAL AND METHOD: This is a retrospective observational study of the results of all exome sequencing performed by the Section of Medical Genetics at Telemark Hospital between December 2012 and October 2014, and includes 125 persons in 46 families. The majority of these families were being investigated for a syndrome (n = 35, 76%) or neurological disease (n = 9, 20%). RESULTS: Exome sequencing detected pathogenic sequence variants in 15 of 46 probands, and variants of unknown significance in 12 probands. Of the 100 patients who stated their wishes regarding feedback of any incidental findings, six indicated that they did not wish to receive such information. There were no incidental findings in this study, but neither were such sequence variants actively looked for. INTERPRETATION: Exome sequencing can enable more patients with syndromes or neurological diseases to receive a causal diagnosis, and to receive this diagnosis at an earlier stage. However, the patients in this study were quite highly selected, and the results must therefore be interpreted with caution.


Subject(s)
Exome , Genetic Diseases, Inborn/diagnosis , High-Throughput Nucleotide Sequencing , Nervous System Diseases , Sequence Analysis, DNA , Humans , Informed Consent , Nervous System Diseases/diagnosis , Nervous System Diseases/genetics , Norway , Retrospective Studies , Syndrome
20.
Hum Mol Genet ; 21(6): 1402-9, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22156580

ABSTRACT

Proprotein convertase subtilisin/kexin type 9 (PCSK9) binds to the epidermal growth factor homology domain repeat A of the low-density lipoprotein receptor (LDLR) at the cell surface and disrupts recycling of the internalized LDLR. As a consequence, the LDLR is rerouted to the lysosomes for degradation. Although PCSK9 may bind to an LDLR lacking the ligand-binding domain, at least three ligand-binding repeats of the ligand-binding domain are required for PCSK9 to reroute the LDLR to the lysosomes. In this study, we have studied the binding of PCSK9 to an LDLR with or without the ligand-binding domain at increasingly acidic conditions in order to mimic the milieu of the LDLR:PCSK9 complex as it translocates from the cell membrane to the sorting endosomes. These studies have shown that PCSK9 is rapidly released from an LDLR lacking the ligand-binding domain at pH in the range of 6.9-6.1. A similar pattern of release at acidic pH was also observed for the binding to the normal LDLR of mutant PCSK9 lacking the C-terminal domain. Together these data indicate that an interaction between the negatively charged ligand-binding domain of the LDLR and the positively charged C-terminal domain of PCSK9 is required for PCSK9 to remain bound to the LDLR during the early phase of endosomal acidification as the LDLR translocates from the cell membrane to the sorting endosome.


Subject(s)
Acids/metabolism , Endosomes/metabolism , Proprotein Convertases/metabolism , Receptors, LDL/metabolism , Serine Endopeptidases/metabolism , Animals , Blotting, Western , CHO Cells , Cell Membrane/metabolism , Cells, Cultured , Cricetinae , Humans , Hydrogen-Ion Concentration , Kidney/cytology , Kidney/metabolism , Lysosomes/metabolism , Mutagenesis, Site-Directed , Mutation/genetics , Proprotein Convertase 9 , Proprotein Convertases/genetics , Protein Binding , Protein Structure, Tertiary , Protein Transport , Receptors, LDL/genetics , Serine Endopeptidases/genetics , Signal Transduction
SELECTION OF CITATIONS
SEARCH DETAIL