Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 58
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Brain ; 146(8): 3513-3527, 2023 08 01.
Article in English | MEDLINE | ID: mdl-36917474

ABSTRACT

RNA polymerase I transcribes ribosomal DNA to produce precursor 47S rRNA. Post-transcriptional processing of this rRNA generates mature 28S, 18S and 5.8S rRNAs, which form the ribosomes, together with 5S rRNA, assembly factors and ribosomal proteins. We previously reported a homozygous variant in the catalytic subunit of RNA polymerase I, POLR1A, in two brothers with leukodystrophy and progressive course. However, the disease mechanism remained unknown. In this report, we describe another missense variant POLR1A NM_015425.3:c.1925C>A; p.(Thr642Asn) in homozygosity in two unrelated patients. Patient 1 was a 16-year-old male and Patient 2 was a 2-year-old female. Both patients manifested neurological deficits, with brain MRIs showing hypomyelinating leukodystrophy and cerebellar atrophy; and in Patient 1 additionally with hypointensity of globi pallidi and small volume of the basal ganglia. Patient 1 had progressive disease course, leading to death at the age of 16.5 years. Extensive in vitro experiments in fibroblasts from Patient 1 documented that the mutated POLR1A led to aberrant rRNA processing and degradation, and abnormal nucleolar homeostasis. Proteomics data analyses and further in vitro experiments documented abnormal protein homeostasis, and endoplasmic reticulum stress responses. We confirm that POLR1A biallelic variants cause neurodegenerative disease, expand the knowledge of the clinical phenotype of the disorder, and provide evidence for possible pathological mechanisms leading to POLR1A-related leukodystrophy.


Subject(s)
Neurodegenerative Diseases , RNA Polymerase I , Male , Female , Humans , RNA Polymerase I/genetics , RNA Polymerase I/metabolism , Neurodegenerative Diseases/genetics , Proteostasis , RNA, Ribosomal/metabolism , Ribosomes , RNA Processing, Post-Transcriptional
2.
Am J Hum Genet ; 106(6): 893-904, 2020 06 04.
Article in English | MEDLINE | ID: mdl-32386558

ABSTRACT

Kinesin-2 enables ciliary assembly and maintenance as an anterograde intraflagellar transport (IFT) motor. Molecular motor activity is driven by a heterotrimeric complex comprised of KIF3A and KIF3B or KIF3C plus one non-motor subunit, KIFAP3. Using exome sequencing, we identified heterozygous KIF3B variants in two unrelated families with hallmark ciliopathy phenotypes. In the first family, the proband presents with hepatic fibrosis, retinitis pigmentosa, and postaxial polydactyly; he harbors a de novo c.748G>C (p.Glu250Gln) variant affecting the kinesin motor domain encoded by KIF3B. The second family is a six-generation pedigree affected predominantly by retinitis pigmentosa. Affected individuals carry a heterozygous c.1568T>C (p.Leu523Pro) KIF3B variant segregating in an autosomal-dominant pattern. We observed a significant increase in primary cilia length in vitro in the context of either of the two mutations while variant KIF3B proteins retained stability indistinguishable from wild type. Furthermore, we tested the effects of KIF3B mutant mRNA expression in the developing zebrafish retina. In the presence of either missense variant, rhodopsin was sequestered to the photoreceptor rod inner segment layer with a concomitant increase in photoreceptor cilia length. Notably, impaired rhodopsin trafficking is also characteristic of recessive KIF3B models as exemplified by an early-onset, autosomal-recessive, progressive retinal degeneration in Bengal cats; we identified a c.1000G>A (p.Ala334Thr) KIF3B variant by genome-wide association study and whole-genome sequencing. Together, our genetic, cell-based, and in vivo modeling data delineate an autosomal-dominant syndromic retinal ciliopathy in humans and suggest that multiple KIF3B pathomechanisms can impair kinesin-driven ciliary transport in the photoreceptor.


Subject(s)
Ciliopathies/genetics , Ciliopathies/pathology , Genes, Dominant/genetics , Kinesins/genetics , Mutation , Retina/pathology , Amino Acid Sequence , Animals , Cats , Child, Preschool , Cilia/pathology , Female , Genome-Wide Association Study , Heterozygote , Humans , Kinesins/chemistry , Kinesins/metabolism , Larva , Male , Middle Aged , Pedigree , Phenotype , Photoreceptor Cells/metabolism , Retina/cytology , Retina/growth & development , Retina/metabolism , Rhodopsin/metabolism , Young Adult , Zebrafish/genetics , Zebrafish/growth & development
3.
Genet Med ; 25(1): 90-102, 2023 01.
Article in English | MEDLINE | ID: mdl-36318270

ABSTRACT

PURPOSE: Brain monoamine vesicular transport disease is an infantile-onset movement disorder that mimics cerebral palsy. In 2013, the homozygous SLC18A2 variant, p.Pro387Leu, was first reported as a cause of this rare disorder, and dopamine agonists were efficient for treating affected individuals from a single large family. To date, only 6 variants have been reported. In this study, we evaluated genotype-phenotype correlations in individuals with biallelic SLC18A2 variants. METHODS: A total of 42 affected individuals with homozygous SLC18A2 variant alleles were identified. We evaluated genotype-phenotype correlations and the missense variants in the affected individuals based on the structural modeling of rat VMAT2 encoded by Slc18a2, with cytoplasm- and lumen-facing conformations. A Caenorhabditis elegans model was created for functional studies. RESULTS: A total of 19 homozygous SLC18A2 variants, including 3 recurrent variants, were identified using exome sequencing. The affected individuals typically showed global developmental delay, hypotonia, dystonia, oculogyric crisis, and autonomic nervous system involvement (temperature dysregulation/sweating, hypersalivation, and gastrointestinal dysmotility). Among the 58 affected individuals described to date, 16 (28%) died before the age of 13 years. Of the 17 patients with p.Pro237His, 9 died, whereas all 14 patients with p.Pro387Leu survived. Although a dopamine agonist mildly improved the disease symptoms in 18 of 21 patients (86%), some affected individuals with p.Ile43Phe and p.Pro387Leu showed milder phenotypes and presented prolonged survival even without treatment. The C. elegans model showed behavioral abnormalities. CONCLUSION: These data expand the phenotypic and genotypic spectra of SLC18A2-related disorders.


Subject(s)
Brain Diseases , Dystonia , Movement Disorders , Humans , Animals , Rats , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Vesicular Monoamine Transport Proteins/genetics , Vesicular Monoamine Transport Proteins/metabolism , Movement Disorders/genetics , Amines , Brain/metabolism
4.
Brain ; 145(7): 2602-2616, 2022 07 29.
Article in English | MEDLINE | ID: mdl-35104841

ABSTRACT

Bi-allelic pathogenic variants in ZBTB11 have been associated with intellectual developmental disorder, autosomal recessive 69 (MRT69; OMIM 618383). We report five patients from three families with novel, bi-allelic variants in ZBTB11. We have expanded the clinical phenotype of MRT69, documenting varied severity of atrophy affecting different brain regions and described combined malonic and methylmalonic aciduria as a biochemical manifestation. As ZBTB11 encodes for a transcriptional regulator, we performeded chromatin immunoprecipitation-sequencing targeting ZBTB11 in fibroblasts from patients and controls. Chromatin immunoprecipitation-sequencing revealed binding of wild-type ZBTB11 to promoters in 238 genes, among which genes encoding proteins involved in mitochondrial functions and RNA processing are over-represented. Mutated ZBTB11 showed reduced binding to 61 of the targeted genes, indicating that the variants act as loss of function. Most of these genes are related to mitochondrial functions. Transcriptome analysis of the patient fibroblasts revealed dysregulation of mitochondrial functions. In addition, we uncovered that reduced binding of the mutated ZBTB11 to ACSF3 leads to decreased ACSF3 transcript level, explaining combined malonic and methylmalonic aciduria. Collectively, these results expand the clinical spectrum of ZBTB11-related neurological disease and give insight into the pathophysiology in which the dysfunctional ZBTB11 affect mitochondrial functions and RNA processing contributing to the neurological and biochemical phenotypes.


Subject(s)
Amino Acid Metabolism, Inborn Errors , Metabolism, Inborn Errors , Nervous System Malformations , Amino Acid Metabolism, Inborn Errors/genetics , Brain , Humans , Metabolism, Inborn Errors/genetics
5.
Hum Mol Genet ; 29(13): 2218-2239, 2020 08 03.
Article in English | MEDLINE | ID: mdl-32504085

ABSTRACT

The RNA exosome is an essential ribonuclease complex required for processing and/or degradation of both coding and non-coding RNAs. We identified five patients with biallelic variants in EXOSC5, which encodes a structural subunit of the RNA exosome. The clinical features of these patients include failure to thrive, short stature, feeding difficulties, developmental delays that affect motor skills, hypotonia and esotropia. Brain MRI revealed cerebellar hypoplasia and ventriculomegaly. While we ascertained five patients, three patients with distinct variants of EXOSC5 were studied in detail. The first patient had a deletion involving exons 5-6 of EXOSC5 and a missense variant, p.Thr114Ile, that were inherited in trans, the second patient was homozygous for p.Leu206His and the third patient had paternal isodisomy for chromosome 19 and was homozygous for p.Met148Thr. The additional two patients ascertained are siblings who had an early frameshift mutation in EXOSC5 and the p.Thr114Ile missense variant that were inherited in trans. We employed three complementary approaches to explore the requirement for EXOSC5 in brain development and assess consequences of pathogenic EXOSC5 variants. Loss of function for exosc5 in zebrafish results in shortened and curved tails/bodies, reduced eye/head size and edema. We modeled pathogenic EXOSC5 variants in both budding yeast and mammalian cells. Some of these variants cause defects in RNA exosome function as well as altered interactions with other RNA exosome subunits. These findings expand the number of genes encoding RNA exosome subunits linked to human disease while also suggesting that disease mechanism varies depending on the specific pathogenic variant.


Subject(s)
Antigens, Neoplasm/genetics , Cerebellum/abnormalities , Developmental Disabilities/genetics , Dwarfism/genetics , Exosome Multienzyme Ribonuclease Complex/genetics , Nervous System Malformations/genetics , RNA-Binding Proteins/genetics , Animals , Cerebellum/pathology , Developmental Disabilities/pathology , Dwarfism/pathology , Frameshift Mutation/genetics , Homozygote , Humans , Mutation, Missense/genetics , Nervous System Malformations/pathology , Pedigree , Zebrafish/genetics , Zebrafish/growth & development
6.
Genet Med ; 24(4): 905-914, 2022 04.
Article in English | MEDLINE | ID: mdl-35027293

ABSTRACT

PURPOSE: Gabriele-de Vries syndrome (GADEVS) is a rare genetic disorder characterized by developmental delay and/or intellectual disability, hypotonia, feeding difficulties, and distinct facial features. To refine the phenotype and to better understand the molecular basis of the syndrome, we analyzed clinical data and performed genome-wide DNA methylation analysis of a series of individuals carrying a YY1 variant. METHODS: Clinical data were collected for 13 individuals not yet reported through an international call for collaboration. DNA was collected for 11 of these individuals and 2 previously reported individuals in an attempt to delineate a specific DNA methylation signature in GADEVS. RESULTS: Phenotype in most individuals overlapped with the previously described features. We described 1 individual with atypical phenotype, heterozygous for a missense variant in a domain usually not involved in individuals with YY1 pathogenic missense variations. We also described a specific peripheral blood DNA methylation profile associated with YY1 variants. CONCLUSION: We reported a distinct DNA methylation episignature in GADEVS. We expanded the clinical profile of GADEVS to include thin/sparse hair and cryptorchidism. We also highlighted the utility of DNA methylation episignature analysis for classification of variants of unknown clinical significance.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , DNA Methylation/genetics , Genome , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Male , Neurodevelopmental Disorders/genetics , Phenotype , Syndrome
7.
Hum Mutat ; 41(10): 1738-1744, 2020 10.
Article in English | MEDLINE | ID: mdl-32643838

ABSTRACT

Congenital heart defects and skeletal malformations syndrome (CHDSKM) is a rare autosomal dominant disorder characterized by congenital heart disease, skeletal abnormalities, and failure to thrive. CHDSKM is caused by germline mutations in ABL1. To date, three variants have been in association with CHDSKM. In this study, we describe three de novo missense variants, c.407C>T (p.Thr136Met), c.746C>T (p.Pro249Leu), and c.1573G>A (p.Val525Met), and one recurrent variant, c.1066G>A (p.Ala356Thr), in six patients, thereby expanding the phenotypic spectrum of CHDSKM to include hearing impairment, lipodystrophy-like features, renal hypoplasia, and distinct ocular abnormalities. Functional investigation of the three novel variants showed an increased ABL1 kinase activity. The cardiac findings in additional patients with p.Ala356Thr contribute to the accumulating evidence that patients carrying either one of the recurrent variants, p.Tyr245Cys and p.Ala356Thr, have a high incidence of cardiac abnormalities. The phenotypic expansion has implications for the clinical diagnosis of CHDSKM in patients with germline ABL1 variants.


Subject(s)
Abnormalities, Multiple , Heart Defects, Congenital , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Germ Cells , Heart Defects, Congenital/genetics , Humans , Phenotype , Syndrome
8.
Hum Mutat ; 41(12): 2179-2194, 2020 12.
Article in English | MEDLINE | ID: mdl-33131181

ABSTRACT

Ciliopathies are clinically and genetically heterogeneous diseases. We studied three patients from two independent families presenting with features of Joubert syndrome: abnormal breathing pattern during infancy, developmental delay/intellectual disability, cerebellar ataxia, molar tooth sign on magnetic resonance imaging scans, and polydactyly. We identified biallelic loss-of-function (LOF) variants in CBY1, segregating with the clinical features of Joubert syndrome in the families. CBY1 localizes to the distal end of the mother centriole, contributing to the formation and function of cilia. In accordance with the clinical and mutational findings in the affected individuals, we demonstrated that depletion of Cby1 in zebrafish causes ciliopathy-related phenotypes. Levels of CBY1 transcript were found reduced in the patients compared with controls, suggesting degradation of the mutated transcript through nonsense-mediated messenger RNA decay. Accordingly, we could detect CBY1 protein in fibroblasts from controls, but not from patients by immunofluorescence. Furthermore, we observed reduced ability to ciliate, increased ciliary length, and reduced levels of the ciliary proteins AHI1 and ARL13B in patient fibroblasts. Our data show that CBY1 LOF-variants cause a ciliopathy with features of Joubert syndrome.


Subject(s)
Abnormalities, Multiple/genetics , Carrier Proteins/genetics , Cerebellum/abnormalities , Ciliopathies/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Mutation/genetics , Nuclear Proteins/genetics , Retina/abnormalities , Abnormalities, Multiple/diagnostic imaging , Abnormalities, Multiple/pathology , Adolescent , Animals , Cerebellum/diagnostic imaging , Cerebellum/pathology , Child , Child, Preschool , Cilia/metabolism , Cilia/pathology , Ciliopathies/diagnostic imaging , Ciliopathies/pathology , Eye Abnormalities/diagnostic imaging , Eye Abnormalities/pathology , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Homozygote , Humans , Infant , Infant, Newborn , Kidney Diseases, Cystic/diagnostic imaging , Kidney Diseases, Cystic/pathology , Magnetic Resonance Imaging , Male , Pedigree , Phenotype , Retina/diagnostic imaging , Retina/pathology , Smoothened Receptor/metabolism , Young Adult , Zebrafish/genetics
9.
Am J Hum Genet ; 100(6): 907-925, 2017 Jun 01.
Article in English | MEDLINE | ID: mdl-28575647

ABSTRACT

Yin and yang 1 (YY1) is a well-known zinc-finger transcription factor with crucial roles in normal development and malignancy. YY1 acts both as a repressor and as an activator of gene expression. We have identified 23 individuals with de novo mutations or deletions of YY1 and phenotypic features that define a syndrome of cognitive impairment, behavioral alterations, intrauterine growth restriction, feeding problems, and various congenital malformations. Our combined clinical and molecular data define "YY1 syndrome" as a haploinsufficiency syndrome. Through immunoprecipitation of YY1-bound chromatin from affected individuals' cells with antibodies recognizing both ends of the protein, we show that YY1 deletions and missense mutations lead to a global loss of YY1 binding with a preferential retention at high-occupancy sites. Finally, we uncover a widespread loss of H3K27 acetylation in particular on the YY1-bound enhancers, underscoring a crucial role for YY1 in enhancer regulation. Collectively, these results define a clinical syndrome caused by haploinsufficiency of YY1 through dysregulation of key transcriptional regulators.


Subject(s)
Chromatin/metabolism , Haploinsufficiency/genetics , Intellectual Disability/genetics , Transcription, Genetic , YY1 Transcription Factor/genetics , Acetylation , Adolescent , Base Sequence , Child, Preschool , Chromatin Immunoprecipitation , Cohort Studies , Enhancer Elements, Genetic/genetics , Female , Gene Ontology , Haplotypes/genetics , Hemizygote , Histones/metabolism , Humans , Lymphocytes/metabolism , Male , Methylation , Models, Molecular , Mutation, Missense/genetics , Protein Binding/genetics , Protein Domains , YY1 Transcription Factor/chemistry
10.
BMC Med Genet ; 21(1): 96, 2020 05 07.
Article in English | MEDLINE | ID: mdl-32381069

ABSTRACT

BACKGROUND: Joubert syndrome (JBTS) is a genetically heterogeneous group of neurodevelopmental syndromes caused by primary cilia dysfunction. Usually the neurological presentation starts with abnormal neonatal breathing followed by muscular hypotonia, psychomotor delay, and cerebellar ataxia. Cerebral MRI shows mid- and hindbrain anomalies including the molar tooth sign. We report a male patient with atypical presentation of Joubert syndrome type 23, thus expanding the phenotype. CASE PRESENTATION: Clinical features were consistent with JBTS already from infancy, yet the syndrome was not suspected before cerebral MRI later in childhood showed the characteristic molar tooth sign and ectopic neurohypophysis. From age 11 years seizures developed and after few years became increasingly difficult to treat, also related to inadequate compliance to therapy. He died at 23 years of sudden unexpected death in epilepsy (SUDEP). The genetic diagnosis remained elusive for many years, despite extensive genetic testing. We reached the genetic diagnosis by performing whole genome sequencing of the family trio and analyzing the data with the combination of one analysis pipeline for single nucleotide variants (SNVs)/indels and one for structural variants (SVs). This lead to the identification of the most common variant detected in patients with JBTS23 (OMIM# 616490), rs534542684, in compound heterozygosity with a 8.3 kb deletion in KIAA0586, not previously reported. CONCLUSIONS: We describe for the first time ectopic neurohypophysis and SUDEP in JBTS23, expanding the phenotype of this condition and raising the attention on the possible severity of the epilepsy in this disease. We also highlight the diagnostic power of WGS, which efficiently detects SNVs/indels and in addition allows the identification of SVs.


Subject(s)
Abnormalities, Multiple/genetics , Cell Cycle Proteins/genetics , Cerebellum/abnormalities , Death, Sudden/pathology , Epilepsy/genetics , Eye Abnormalities/genetics , Kidney Diseases, Cystic/genetics , Retina/abnormalities , Abnormalities, Multiple/mortality , Abnormalities, Multiple/pathology , Adult , Cerebellum/pathology , Child , Death, Sudden/epidemiology , Developmental Disabilities/genetics , Developmental Disabilities/mortality , Developmental Disabilities/pathology , Epilepsy/mortality , Epilepsy/pathology , Eye Abnormalities/mortality , Eye Abnormalities/pathology , Female , Heterozygote , Humans , INDEL Mutation , Kidney Diseases, Cystic/mortality , Kidney Diseases, Cystic/pathology , Male , Pituitary Gland, Posterior/metabolism , Pituitary Gland, Posterior/pathology , Retina/pathology , Whole Genome Sequencing , Young Adult
11.
Genet Med ; 20(7): 778-784, 2018 07.
Article in English | MEDLINE | ID: mdl-28837161

ABSTRACT

PURPOSE: To elucidate the novel molecular cause in two unrelated consanguineous families with autosomal recessive intellectual disability. METHODS: A combination of homozygosity mapping and exome sequencing was used to locate the plausible genetic defect in family F162, while only exome sequencing was followed in the family PKMR65. The protein 3D structure was visualized with the University of California-San Francisco Chimera software. RESULTS: All five patients from both families presented with severe intellectual disability, aggressive behavior, and speech and motor delay. Four of the five patients had microcephaly. We identified homozygous missense variants in LINGO1, p.(Arg290His) in family F162 and p.(Tyr288Cys) in family PKMR65. Both variants were predicted to be pathogenic, and segregated with the phenotype in the respective families. Molecular modeling of LINGO1 suggests that both variants interfere with the glycosylation of the protein. CONCLUSION: LINGO1 is a transmembrane receptor, predominantly found in the central nervous system. Published loss-of-function studies in mouse and zebrafish have established a crucial role of LINGO1 in normal neuronal development and central nervous system myelination by negatively regulating oligodendrocyte differentiation and neuronal survival. Taken together, our results indicate that biallelic LINGO1 missense variants cause autosomal recessive intellectual disability in humans.


Subject(s)
Intellectual Disability/genetics , Membrane Proteins/genetics , Nerve Tissue Proteins/genetics , Alleles , Chromosome Mapping/methods , Family , Female , Gene Frequency/genetics , Genotype , Homozygote , Humans , Language Development Disorders/genetics , Male , Membrane Proteins/physiology , Microcephaly/genetics , Motor Activity/genetics , Mutation, Missense/genetics , Nerve Tissue Proteins/physiology , Pakistan , Pedigree , Phenotype , Sequence Analysis, Protein , Exome Sequencing
12.
J Med Genet ; 54(7): 460-470, 2017 07.
Article in English | MEDLINE | ID: mdl-28377535

ABSTRACT

BACKGROUND: We aimed for a comprehensive delineation of genetic, functional and phenotypic aspects of GRIN2B encephalopathy and explored potential prospects of personalised medicine. METHODS: Data of 48 individuals with de novo GRIN2B variants were collected from several diagnostic and research cohorts, as well as from 43 patients from the literature. Functional consequences and response to memantine treatment were investigated in vitro and eventually translated into patient care. RESULTS: Overall, de novo variants in 86 patients were classified as pathogenic/likely pathogenic. Patients presented with neurodevelopmental disorders and a spectrum of hypotonia, movement disorder, cortical visual impairment, cerebral volume loss and epilepsy. Six patients presented with a consistent malformation of cortical development (MCD) intermediate between tubulinopathies and polymicrogyria. Missense variants cluster in transmembrane segments and ligand-binding sites. Functional consequences of variants were diverse, revealing various potential gain-of-function and loss-of-function mechanisms and a retained sensitivity to the use-dependent blocker memantine. However, an objectifiable beneficial treatment response in the respective patients still remains to be demonstrated. CONCLUSIONS: In addition to previously known features of intellectual disability, epilepsy and autism, we found evidence that GRIN2B encephalopathy is also frequently associated with movement disorder, cortical visual impairment and MCD revealing novel phenotypic consequences of channelopathies.


Subject(s)
Brain Diseases/genetics , Mutation/genetics , Receptors, N-Methyl-D-Aspartate/genetics , Brain Diseases/drug therapy , Heterozygote , Humans , Magnetic Resonance Imaging , Memantine/therapeutic use , Molecular Targeted Therapy , Neuroimaging , Phenotype , Receptors, N-Methyl-D-Aspartate/antagonists & inhibitors , Receptors, N-Methyl-D-Aspartate/metabolism
13.
Hum Mol Genet ; 24(20): 5845-54, 2015 Oct 15.
Article in English | MEDLINE | ID: mdl-26220973

ABSTRACT

Import of peroxisomal matrix proteins, crucial for peroxisome biogenesis, is mediated by the cytosolic receptors PEX5 and PEX7 that recognize proteins carrying peroxisomal targeting signals 1 or 2 (PTS1 or PTS2), respectively. Mutations in PEX5 or 12 other PEX genes cause peroxisome biogenesis disorders, collectively named the Zellweger spectrum disorders (ZSDs), whereas mutations in PEX7 cause rhizomelic chondrodysplasia punctata type 1 (RCDP1). Three additional RCDP types, RCDP2-3-4, are caused, respectively, by mutations in GNPAT, AGPS and FAR1, encoding enzymes involved in plasmalogen biosynthesis. Here we report a fifth type of RCDP (RCDP5) caused by a novel mutation in PEX5. In four patients with RCDP from two independent families, we identified a homozygous frame shift mutation c.722dupA (p.Val242Glyfs(∗)33) in PEX5 (GenBank: NM_001131023.1). PEX5 encodes two isoforms, PEX5L and PEX5S, and we show that the c.722dupA mutation, located in the PEX5L-specific exon 9, results in loss of PEX5L only. Both PEX5 isoforms recognize PTS1-tagged proteins, but PEX5L is also a co-receptor for PTS2-tagged proteins. Previous patients with PEX5 mutations had ZSD, mainly due to deficient import of PTS1-tagged proteins. Similarly to mutations in PEX7, loss of PEX5L results in deficient import of PTS2-tagged proteins only, thus causing RCDP instead of ZSD. We demonstrate that PEX5L expression restores the import of PTS2-tagged proteins in patient fibroblasts. Due to the biochemical overlap between RCDP1 and RCDP5, sequencing of PEX7 and exon 9 in PEX5 should be performed in patients with a selective defect in the import of PTS2-tagged proteins.


Subject(s)
Chondrodysplasia Punctata, Rhizomelic/genetics , Frameshift Mutation , Peroxisomes/metabolism , Protein Transport/genetics , Receptors, Cytoplasmic and Nuclear/genetics , Adolescent , Adult , Child , Chondrodysplasia Punctata, Rhizomelic/metabolism , Exome , Female , Humans , Infant , Male , Pedigree , Peroxisome-Targeting Signal 1 Receptor , Peroxisomes/genetics , Protein Isoforms , Receptors, Cytoplasmic and Nuclear/metabolism , Sequence Analysis, DNA
14.
BMC Med Genet ; 16: 113, 2015 Dec 18.
Article in English | MEDLINE | ID: mdl-26684006

ABSTRACT

BACKGROUND: Pathogenic mutations in FBN1, encoding the glycoprotein, fibrillin-1, cause Marfan syndrome (MFS) and related connective tissue disorders. In the present study, qualitative and quantitative effects of 16 mutations, identified in FBN1 in MFS patients with systematically described phenotypes, were investigated in vitro. METHODS: Qualitative analysis was performed with reverse transcription-PCR (RT-PCR) and gel electrophoresis, and quantitative analysis to determine the FBN1 mRNA levels in fibroblasts from the 16 patients with MFS was performed with real-time PCR. RESULTS: Qualitative analysis documented that the mutations c.4817-2delA and c.A4925G led to aberrant FBN1 mRNA splicing leading to in frame deletion of exon 39 and in exon 39, respectively. No difference in the mean FBN1 mRNA level was observed between the entire group of cases and controls, nor between the group of patients with missense mutations and controls. The mean expression levels associated with premature termination codon (PTC) and splice site mutations were significantly lower than the levels in patients with missense mutations. A high level of FBN1 mRNA in the patient with the missense mutation c.G2447T did not segregate with the mutation in three of his first degree relatives. No association was indicated between the FBN1 transcript level and specific phenotypic manifestations. CONCLUSIONS: Abnormal FBN1 transcripts were indicated in fibroblasts from patients with the splice site mutation c.4817-2delA and the missense mutation c.A4925G. While the mean FBN1 mRNA expression level in fibroblasts from patients with splice site and PTC mutations were lower than the mean level in patients with missense mutations and controls, inter-individual variability was high. The observation that high level of FBN1 mRNA in the patient with the missense mutation c.G2447T did not segregate with the mutation in the family suggests that variable expression of the normal FBN1 allele may contribute to explain the variability in FBN1 mRNA level.


Subject(s)
Fibroblasts/metabolism , Marfan Syndrome/genetics , Microfilament Proteins/genetics , RNA, Messenger/genetics , Base Sequence , Cells, Cultured , DNA Mutational Analysis , Fibrillin-1 , Fibrillins , Genetic Predisposition to Disease/genetics , Genotype , Humans , Marfan Syndrome/metabolism , Marfan Syndrome/pathology , Mutation , Mutation, Missense , RNA Splice Sites/genetics , RNA Splicing/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sequence Deletion
15.
Am J Med Genet A ; 167A(8): 1890-6, 2015 Aug.
Article in English | MEDLINE | ID: mdl-25846056

ABSTRACT

We report on a 10-year-old-boy presenting with moderate intellectual disability (ID), impaired motor skills, hypotonia, growth delay, minor anomalies, misaligned teeth, pectus excavatum, small hands and feet, widely spaced nipples, and a 1.13 Mb de novo deletion on HSA12q12 (chr12:44,830,147-45,964,945 bp, hg19), deleting ANO6, NELL2, and DBX2 and the pseudogenes PLEKHA8P1 and RACGAP1P. We suggest DBX2 and NELL2 as disease-causing genes and their haploinsufficiency to be involved in the psychomotor delay in the patient. DBX2 encodes a homeobox protein, highly expressed during neuronal development and regulating differentiation of interneurons in brain and spinal cord. NELL2 is expressed in most of the central and peripheral nervous system, with highest expression in hippocampus and cerebellum, maximizing during neuronal differentiation. The deletion in our patient is the smallest in HSA12q12 reported to date, and it is included in the deletion carried by four previously reported patients. The clinical presentation of these patients points to the recurrence of the following manifestation, possibly delineating a 12q12 deletion syndrome phenotype: moderate to severe developmental/intellectual delay, hypotonia, postnatal growth retardation, skeletal and dental anomalies, minor facial anomalies including strabismus, down slanting palpebral fissures, and large/low-set ears.


Subject(s)
Developmental Disabilities/genetics , Haploinsufficiency , Intellectual Disability/genetics , Nerve Tissue Proteins/genetics , Phospholipid Transfer Proteins/genetics , Anoctamins , Child , Humans , Male
16.
Am J Med Genet A ; 167A(3): 657-63, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25691420

ABSTRACT

A pair of sisters was ascertained for multiple congenital defects, including marked craniofacial dysmorphisms with blepharophimosis, and severe psychomotor delay. Two novel compound heterozygous mutations in UBE3B were identified in both the sisters by exome sequencing. These mutations include c.1A>G, which predicts p.Met1?, and a c.1773delC variant, predicted to cause a frameshift at p.Phe591fs. UBE3B encodes a widely expressed protein ubiquitin ligase E3B, which, when mutated in both alleles, causes Kaufman oculocerebrofacial syndrome. We report on the thorough clinical examination of the patients and review the state of art knowledge of this disorder.


Subject(s)
Eye Abnormalities/diagnosis , Eye Abnormalities/genetics , Heterozygote , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Limb Deformities, Congenital/diagnosis , Limb Deformities, Congenital/genetics , Microcephaly/diagnosis , Microcephaly/genetics , Mutation , Phenotype , Ubiquitin-Protein Ligases/genetics , Child, Preschool , Comparative Genomic Hybridization , DNA Mutational Analysis , Exome , Facies , Female , Genetic Association Studies , High-Throughput Nucleotide Sequencing , Humans , Infant , Pedigree
17.
Hum Mutat ; 35(5): 556-64, 2014 May.
Article in English | MEDLINE | ID: mdl-24619930

ABSTRACT

Stormorken syndrome is a rare autosomal-dominant disease with mild bleeding tendency, thrombocytopathy, thrombocytopenia, mild anemia, asplenia, tubular aggregate myopathy, miosis, headache, and ichthyosis. A heterozygous missense mutation in STIM1 exon 7 (c.910C>T; p.Arg304Trp) (NM_003156.3) was found to segregate with the disease in six Stormorken syndrome patients in four families. Upon sensing Ca(2+) depletion in the endoplasmic reticulum lumen, STIM1 undergoes a conformational change enabling it to interact with and open ORAI1, a Ca(2+) release-activated Ca(2+) channel located in the plasma membrane. The STIM1 mutation found in Stormorken syndrome patients is located in the coiled-coil 1 domain, which might play a role in keeping STIM1 inactive. In agreement with a possible gain-of-function mutation in STIM1, blood platelets from patients were in a preactivated state with high exposure of aminophospholipids on the outer surface of the plasma membrane. Resting Ca(2+) levels were elevated in platelets from the patients compared with controls, and store-operated Ca(2+) entry was markedly attenuated, further supporting constitutive activity of STIM1 and ORAI1. Thus, our data are compatible with a near-maximal activation of STIM1 in Stormorken syndrome patients. We conclude that the heterozygous mutation c.910C>T causes the complex phenotype that defines this syndrome.


Subject(s)
Blood Platelet Disorders/genetics , Dyslexia/genetics , Exome/genetics , Ichthyosis/genetics , Membrane Proteins/genetics , Migraine Disorders/genetics , Miosis/genetics , Mutation, Missense/genetics , Neoplasm Proteins/genetics , Spleen/abnormalities , Adult , Blood Platelet Disorders/pathology , Calcium Channels/genetics , Comparative Genomic Hybridization , Dyslexia/pathology , Erythrocytes, Abnormal/pathology , Female , High-Throughput Nucleotide Sequencing , Humans , Ichthyosis/pathology , Male , Migraine Disorders/pathology , Miosis/pathology , Muscle Fatigue/genetics , ORAI1 Protein , Pedigree , Spleen/pathology , Stromal Interaction Molecule 1
18.
Genes (Basel) ; 15(4)2024 04 17.
Article in English | MEDLINE | ID: mdl-38674434

ABSTRACT

Oxidative phosphorylation involves a complex multi-enzymatic mitochondrial machinery critical for proper functioning of the cell, and defects herein cause a wide range of diseases called "primary mitochondrial disorders" (PMDs). Mutations in about 400 nuclear and 37 mitochondrial genes have been documented to cause PMDs, which have an estimated birth prevalence of 1:5000. Here, we describe a 4-year-old female presenting from early childhood with psychomotor delay and white matter signal changes affecting several brain regions, including the brainstem, in addition to lactic and phytanic acidosis, compatible with Leigh syndrome, a genetically heterogeneous subgroup of PMDs. Whole genome sequencing of the family trio identified a homozygous 12.9 Kb deletion, entirely overlapping the NDUFA4 gene. Sanger sequencing of the breakpoints revealed that the genomic rearrangement was likely triggered by Alu elements flanking the gene. NDUFA4 encodes for a subunit of the respiratory chain Complex IV, whose activity was significantly reduced in the patient's fibroblasts. In one family, dysfunction of NDUFA4 was previously documented as causing mitochondrial Complex IV deficiency nuclear type 21 (MC4DN21, OMIM 619065), a relatively mild form of Leigh syndrome. Our finding confirms the loss of NDUFA4 function as an ultra-rare cause of Complex IV defect, clinically presenting as Leigh syndrome.


Subject(s)
Electron Transport Complex I , Leigh Disease , Humans , Leigh Disease/genetics , Leigh Disease/pathology , Female , Child, Preschool , Electron Transport Complex IV/genetics , Mitochondrial Diseases/genetics , Mitochondrial Diseases/pathology , Pedigree , Sequence Deletion
19.
Commun Biol ; 7(1): 831, 2024 Jul 08.
Article in English | MEDLINE | ID: mdl-38977784

ABSTRACT

Microtubule associated proteins (MAPs) are widely expressed in the central nervous system, and have established roles in cell proliferation, myelination, neurite formation, axon specification, outgrowth, dendrite, and synapse formation. We report eleven individuals from seven families harboring predicted pathogenic biallelic, de novo, and heterozygous variants in the NAV3 gene, which encodes the microtubule positive tip protein neuron navigator 3 (NAV3). All affected individuals have intellectual disability (ID), microcephaly, skeletal deformities, ocular anomalies, and behavioral issues. In mouse brain, Nav3 is expressed throughout the nervous system, with more prominent signatures in postmitotic, excitatory, inhibiting, and sensory neurons. When overexpressed in HEK293T and COS7 cells, pathogenic variants impaired NAV3 ability to stabilize microtubules. Further, knocking-down nav3 in zebrafish led to severe morphological defects, microcephaly, impaired neuronal growth, and behavioral impairment, which were rescued with co-injection of WT NAV3 mRNA and not by transcripts encoding the pathogenic variants. Our findings establish the role of NAV3 in neurodevelopmental disorders, and reveal its involvement in neuronal morphogenesis, and neuromuscular responses.


Subject(s)
Developmental Disabilities , Intellectual Disability , Microcephaly , Animals , Child , Child, Preschool , Female , Humans , Male , Mice , Chlorocebus aethiops , COS Cells , Developmental Disabilities/genetics , HEK293 Cells , Intellectual Disability/genetics , Microcephaly/genetics , Microcephaly/pathology , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Nerve Tissue Proteins/genetics , Nerve Tissue Proteins/metabolism , Neurons/metabolism , Neurons/pathology , Zebrafish/genetics
20.
Am J Med Genet A ; 161A(5): 1137-42, 2013 May.
Article in English | MEDLINE | ID: mdl-23463539

ABSTRACT

The transcription factor SOX3 is widely expressed in early vertebrate brain development. In humans, duplication of SOX3 and polyalanine expansions at its C-terminus may cause intellectual disability and hypopituitarism. Sox3 knock-out mice show a variable phenotype including structural and functional anomalies affecting the branchial arches and midline cerebral structures such as the optic chiasm and the hypothalamo-pituitary axis. SOX3 is claimed to be required in normal brain development and function in mice and humans, as well as in pituitary and craniofacial development. We report on an 8-year-old boy with a 2.1 Mb deletion in Xq27.1q27.2, which was found to be inherited from his healthy mother. To our knowledge, this is the smallest deletion including the entire SOX3 gene in a male reported to date. He is mildly intellectually disabled with language delay, dysarthria, behavior problems, minor facial anomalies, and hyperphagia. Hormone levels including growth, adrenocorticotropic and thyroid stimulating hormones are normal. Magnetic resonance imaging (MRI) at age 6 years showed no obvious brain anomalies. Genetic redundancy between the three members of the B1 subfamily of SOX proteins during early human brain development likely explains the apparently normal development of brain structures in our patient who is nullisomic for SOX3.


Subject(s)
Brain/abnormalities , Developmental Disabilities/genetics , Hyperphagia/genetics , Intellectual Disability/genetics , SOXB1 Transcription Factors/genetics , Child , Cytogenetic Analysis , Humans , Male , Polymerase Chain Reaction , Sequence Deletion
SELECTION OF CITATIONS
SEARCH DETAIL