Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 35
Filter
1.
Proc Natl Acad Sci U S A ; 119(26): e2204084119, 2022 06 28.
Article in English | MEDLINE | ID: mdl-35727972

ABSTRACT

Discovery of deafness genes and elucidating their functions have substantially contributed to our understanding of hearing physiology and its pathologies. Here we report on DNA variants in MINAR2, encoding membrane integral NOTCH2-associated receptor 2, in four families underlying autosomal recessive nonsyndromic deafness. Neurologic evaluation of affected individuals at ages ranging from 4 to 80 y old does not show additional abnormalities. MINAR2 is a recently annotated gene with limited functional understanding. We detected three MINAR2 variants, c.144G > A (p.Trp48*), c.412_419delCGGTTTTG (p.Arg138Valfs*10), and c.393G > T, in 13 individuals with congenital- or prelingual-onset severe-to-profound sensorineural hearing loss (HL). The c.393G > T variant is shown to disrupt a splice donor site. We show that Minar2 is expressed in the mouse inner ear, with the protein localizing mainly in the hair cells, spiral ganglia, the spiral limbus, and the stria vascularis. Mice with loss of function of the Minar2 protein (Minar2tm1b/tm1b) present with rapidly progressive sensorineural HL associated with a reduction in outer hair cell stereocilia in the shortest row and degeneration of hair cells at a later age. We conclude that MINAR2 is essential for hearing in humans and mice and its disruption leads to sensorineural HL. Progressive HL observed in mice and in some affected individuals and as well as relative preservation of hair cells provides an opportunity to interfere with HL using genetic therapies.


Subject(s)
Hearing Loss, Sensorineural , Receptor, Notch2 , Receptors, Cell Surface , Animals , Hearing Loss, Sensorineural/genetics , Humans , Loss of Function Mutation , Mice , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptors, Cell Surface/genetics , Stereocilia/metabolism
2.
PLoS Genet ; 17(9): e1009803, 2021 09.
Article in English | MEDLINE | ID: mdl-34570759

ABSTRACT

SNIP1 (Smad nuclear interacting protein 1) is a widely expressed transcriptional suppressor of the TGF-ß signal-transduction pathway which plays a key role in human spliceosome function. Here, we describe extensive genetic studies and clinical findings of a complex inherited neurodevelopmental disorder in 35 individuals associated with a SNIP1 NM_024700.4:c.1097A>G, p.(Glu366Gly) variant, present at high frequency in the Amish community. The cardinal clinical features of the condition include hypotonia, global developmental delay, intellectual disability, seizures, and a characteristic craniofacial appearance. Our gene transcript studies in affected individuals define altered gene expression profiles of a number of molecules with well-defined neurodevelopmental and neuropathological roles, potentially explaining clinical outcomes. Together these data confirm this SNIP1 gene variant as a cause of an autosomal recessive complex neurodevelopmental disorder and provide important insight into the molecular roles of SNIP1, which likely explain the cardinal clinical outcomes in affected individuals, defining potential therapeutic avenues for future research.


Subject(s)
Alleles , Amish/genetics , Neurodevelopmental Disorders/genetics , RNA-Binding Proteins/genetics , Gene Expression/genetics , Genes, Recessive , Humans
3.
Genome Res ; 29(7): 1057-1066, 2019 07.
Article in English | MEDLINE | ID: mdl-31160375

ABSTRACT

Germline mutations in fundamental epigenetic regulatory molecules including DNA methyltransferase 3 alpha (DNMT3A) are commonly associated with growth disorders, whereas somatic mutations are often associated with malignancy. We profiled genome-wide DNA methylation patterns in DNMT3A c.2312G > A; p.(Arg771Gln) carriers in a large Amish sibship with Tatton-Brown-Rahman syndrome (TBRS), their mosaic father, and 15 TBRS patients with distinct pathogenic de novo DNMT3A variants. This defined widespread DNA hypomethylation at specific genomic sites enriched at locations annotated as genes involved in morphogenesis, development, differentiation, and malignancy predisposition pathways. TBRS patients also displayed highly accelerated DNA methylation aging. These findings were most marked in a carrier of the AML-associated driver mutation p.Arg882Cys. Our studies additionally defined phenotype-related accelerated and decelerated epigenetic aging in two histone methyltransferase disorders: NSD1 Sotos syndrome overgrowth disorder and KMT2D Kabuki syndrome growth impairment. Together, our findings provide fundamental new insights into aberrant epigenetic mechanisms, the role of epigenetic machinery maintenance, and determinants of biological aging in these growth disorders.


Subject(s)
Aging/genetics , DNA (Cytosine-5-)-Methyltransferases/genetics , Epigenesis, Genetic , Growth Disorders/genetics , Mutation , Abnormalities, Multiple/genetics , Adolescent , Adult , Amish/genetics , Child , DNA Methylation , DNA Methyltransferase 3A , Face/abnormalities , Hematologic Diseases/genetics , Humans , Intellectual Disability/genetics , Leukemia, Myeloid, Acute/genetics , Male , Methyltransferases , Morphogenesis/genetics , Syndrome , Vestibular Diseases/genetics , Young Adult
4.
Genet Med ; 24(3): 631-644, 2022 03.
Article in English | MEDLINE | ID: mdl-34906488

ABSTRACT

PURPOSE: We previously defined biallelic HYAL2 variants causing a novel disorder in 2 families, involving orofacial clefting, facial dysmorphism, congenital heart disease, and ocular abnormalities, with Hyal2 knockout mice displaying similar phenotypes. In this study, we better define the phenotype and pathologic disease mechanism. METHODS: Clinical and genomic investigations were undertaken alongside molecular studies, including immunoblotting and immunofluorescence analyses of variant/wild-type human HYAL2 expressed in mouse fibroblasts, and in silico modeling of putative pathogenic variants. RESULTS: Ten newly identified individuals with this condition were investigated, and they were associated with 9 novel pathogenic variants. Clinical studies defined genotype-phenotype correlations and confirmed a recognizable craniofacial phenotype in addition to myopia, cleft lip/palate, and congenital cardiac anomalies as the most consistent manifestations of the condition. In silico modeling of missense variants identified likely deleterious effects on protein folding. Consistent with this, functional studies indicated that these variants cause protein instability and a concomitant cell surface absence of HYAL2 protein. CONCLUSION: These studies confirm an association between HYAL2 alterations and syndromic cleft lip/palate, provide experimental evidence for the pathogenicity of missense alleles, enable further insights into the pathomolecular basis of the disease, and delineate the core and variable clinical outcomes of the condition.


Subject(s)
Cleft Lip , Cleft Palate , Alleles , Animals , Cell Adhesion Molecules/genetics , Cleft Lip/genetics , Cleft Palate/genetics , GPI-Linked Proteins/genetics , Genetic Association Studies , Humans , Hyaluronoglucosaminidase/genetics , Mice , Phenotype
5.
Am J Med Genet A ; 188(2): 498-508, 2022 02.
Article in English | MEDLINE | ID: mdl-34697879

ABSTRACT

Autosomal recessive limb-girdle muscular dystrophy-1 (LGMDR1) is an autosomal recessive disorder characterized by progressive weakness of the proximal limb and girdle muscles. Biallelic mutations in CAPN3 are reported frequently to cause LGMDR1. Here, we describe 11 individuals from three unrelated consanguineous families that present with typical features of LGMDR1 that include proximal muscle wasting, weakness of the upper and lower limbs, and elevated serum creatine kinase. Whole-exome sequencing identified a rare homozygous CAPN3 variant near the exon 2 splice donor site that segregates with disease in all three families. mRNA splicing studies showed partial retention of intronic sequence and subsequent introduction of a premature stop codon (NM_000070.3: c.379 + 3A>G; p.Asp128Glyfs*15). Furthermore, we observe reduced CAPN3 expression in primary dermal fibroblasts derived from an affected individual, suggesting instability and/or nonsense-mediated decay of mutation-bearing mRNA. Genome-wide homozygosity mapping and single-nucleotide polymorphism analysis identified a shared haplotype and supports a possible founder effect for the CAPN3 variant. Together, our data extend the mutational spectrum of LGMDR1 and have implications for improved diagnostics for individuals of Pakistani origin.


Subject(s)
Calpain , Muscular Dystrophies, Limb-Girdle , Calpain/genetics , Humans , Muscle Proteins/genetics , Muscular Dystrophies, Limb-Girdle/diagnosis , Muscular Dystrophies, Limb-Girdle/genetics , Mutation , Pakistan , RNA, Messenger/genetics
6.
Brain ; 144(12): 3597-3610, 2021 12 31.
Article in English | MEDLINE | ID: mdl-34415310

ABSTRACT

Phosphatidylinositol 4-kinase IIIα (PI4KIIIα/PI4KA/OMIM:600286) is a lipid kinase generating phosphatidylinositol 4-phosphate (PI4P), a membrane phospholipid with critical roles in the physiology of multiple cell types. PI4KIIIα's role in PI4P generation requires its assembly into a heterotetrameric complex with EFR3, TTC7 and FAM126. Sequence alterations in two of these molecular partners, TTC7 (encoded by TTC7A or TCC7B) and FAM126, have been associated with a heterogeneous group of either neurological (FAM126A) or intestinal and immunological (TTC7A) conditions. Here we show that biallelic PI4KA sequence alterations in humans are associated with neurological disease, in particular hypomyelinating leukodystrophy. In addition, affected individuals may present with inflammatory bowel disease, multiple intestinal atresia and combined immunodeficiency. Our cellular, biochemical and structural modelling studies indicate that PI4KA-associated phenotypical outcomes probably stem from impairment of PI4KIIIα-TTC7-FAM126's organ-specific functions, due to defective catalytic activity or altered intra-complex functional interactions. Together, these data define PI4KA gene alteration as a cause of a variable phenotypical spectrum and provide fundamental new insight into the combinatorial biology of the PI4KIIIα-FAM126-TTC7-EFR3 molecular complex.


Subject(s)
Hereditary Central Nervous System Demyelinating Diseases/genetics , Intestinal Atresia/genetics , Minor Histocompatibility Antigens/genetics , Phosphotransferases (Alcohol Group Acceptor)/genetics , Primary Immunodeficiency Diseases/genetics , Female , Humans , Male , Pedigree , Polymorphism, Single Nucleotide
7.
PLoS Genet ; 14(8): e1007504, 2018 08.
Article in English | MEDLINE | ID: mdl-30157172

ABSTRACT

We identified a homozygous missense alteration (c.75C>A, p.D25E) in CLCC1, encoding a presumptive intracellular chloride channel highly expressed in the retina, associated with autosomal recessive retinitis pigmentosa (arRP) in eight consanguineous families of Pakistani descent. The p.D25E alteration decreased CLCC1 channel function accompanied by accumulation of mutant protein in granules within the ER lumen, while siRNA knockdown of CLCC1 mRNA induced apoptosis in cultured ARPE-19 cells. TALEN KO in zebrafish was lethal 11 days post fertilization. The depressed electroretinogram (ERG) cone response and cone spectral sensitivity of 5 dpf KO zebrafish and reduced eye size, retinal thickness, and expression of rod and cone opsins could be rescued by injection of wild type CLCC1 mRNA. Clcc1+/- KO mice showed decreased ERGs and photoreceptor number. Together these results strongly suggest that intracellular chloride transport by CLCC1 is a critical process in maintaining retinal integrity, and CLCC1 is crucial for survival and function of retinal cells.


Subject(s)
Chloride Channels/genetics , Mutation, Missense , Retinitis Pigmentosa/genetics , Animals , Asian People/genetics , Cell Line , Chloride Channels/metabolism , Cytoplasm/metabolism , Eye Proteins/genetics , Eye Proteins/metabolism , HEK293 Cells , Homozygote , Humans , Mice , Mice, Knockout , Pakistan , Retina/metabolism , Retinal Cone Photoreceptor Cells/metabolism , Retinal Rod Photoreceptor Cells/metabolism , Retinitis Pigmentosa/diagnosis , Zebrafish/genetics , Zebrafish/metabolism
8.
PLoS Genet ; 13(1): e1006470, 2017 Jan.
Article in English | MEDLINE | ID: mdl-28081210

ABSTRACT

Orofacial clefting is amongst the most common of birth defects, with both genetic and environmental components. Although numerous studies have been undertaken to investigate the complexities of the genetic etiology of this heterogeneous condition, this factor remains incompletely understood. Here, we describe mutations in the HYAL2 gene as a cause of syndromic orofacial clefting. HYAL2, encoding hyaluronidase 2, degrades extracellular hyaluronan, a critical component of the developing heart and palatal shelf matrix. Transfection assays demonstrated that the gene mutations destabilize the molecule, dramatically reducing HYAL2 protein levels. Consistent with the clinical presentation in affected individuals, investigations of Hyal2-/- mice revealed craniofacial abnormalities, including submucosal cleft palate. In addition, cor triatriatum sinister and hearing loss, identified in a proportion of Hyal2-/- mice, were also found as incompletely penetrant features in affected humans. Taken together our findings identify a new genetic cause of orofacial clefting in humans and mice, and define the first molecular cause of human cor triatriatum sinister, illustrating the fundamental importance of HYAL2 and hyaluronan turnover for normal human and mouse development.


Subject(s)
Cell Adhesion Molecules/genetics , Cleft Lip/genetics , Cleft Palate/genetics , Cor Triatriatum/genetics , Hyaluronoglucosaminidase/genetics , Mutation , Adolescent , Animals , Child , Child, Preschool , Cleft Lip/pathology , Cleft Palate/pathology , Cor Triatriatum/pathology , Female , GPI-Linked Proteins/genetics , Humans , Male , Mice , Mice, Inbred C57BL , Pedigree , Penetrance , Syndrome
9.
BMC Med Genet ; 19(1): 25, 2018 02 20.
Article in English | MEDLINE | ID: mdl-29458334

ABSTRACT

BACKGROUND: L-2-hydroxyglutaric aciduria (L2HGA) is a progressive neurometabolic disease of brain caused by mutations of in L-2-hydroxyglutarate dehydrogenase (L2HGDH) gene. Cardinal clinical features include cerebellar ataxia, epilepsy, neurodevelopmental delay, intellectual disability, and other clinical neurological deficits. CASE PRESENTATION: We describe an index case of the family presented with generalised tonic-clonic seizure, developmental delay, intellectual disability, and ataxia. Initially, the differential diagnosis was difficult to be established and a SNP genome wide scan identified the candidate region on chromosome 14q22.1. DNA sequencing showed a novel homozygous mutation in the candidate gene L2HGDH (NM_024884.2: c.178G > A; p.Gly60Arg). The mutation p.Gly60Arg lies in the highly conserved FAD/NAD(P)-binding domain of this mitochondrial enzyme, predicted to disturb enzymatic function. CONCLUSIONS: The combination of homozygosity mapping and DNA sequencing identified a novel mutation in Pakistani family with variable clinical features. This is second report of a mutation in L2HGDH gene from Pakistan and the largest family with L2HGA reported to date.


Subject(s)
Alcohol Oxidoreductases/genetics , Ataxia/genetics , Consanguinity , Epilepsy/genetics , Intellectual Disability/genetics , Seizures/genetics , Adolescent , Amino Acid Sequence , Asian People/genetics , Ataxia/diagnosis , Chromosome Mapping , Chromosomes, Human, Pair 14/genetics , Computational Biology , Epilepsy/diagnosis , Female , Homozygote , Humans , Intellectual Disability/diagnosis , Mutation , Mutation, Missense , Pakistan , Pedigree , Polymorphism, Single Nucleotide , Protein Conformation , Seizures/diagnosis , Sequence Analysis, DNA
10.
Brain ; 140(11): 2838-2850, 2017 Nov 01.
Article in English | MEDLINE | ID: mdl-29088354

ABSTRACT

The presynaptic, high-affinity choline transporter is a critical determinant of signalling by the neurotransmitter acetylcholine at both central and peripheral cholinergic synapses, including the neuromuscular junction. Here we describe an autosomal recessive presynaptic congenital myasthenic syndrome presenting with a broad clinical phenotype due to homozygous choline transporter missense mutations. The clinical phenotype ranges from the classical presentation of a congenital myasthenic syndrome in one patient (p.Pro210Leu), to severe neurodevelopmental delay with brain atrophy (p.Ser94Arg) and extend the clinical outcomes to a more severe spectrum with infantile lethality (p.Val112Glu). Cells transfected with mutant transporter construct revealed a virtually complete loss of transport activity that was paralleled by a reduction in transporter cell surface expression. Consistent with these findings, studies to determine the impact of gene mutations on the trafficking of the Caenorhabditis elegans choline transporter orthologue revealed deficits in transporter export to axons and nerve terminals. These findings contrast with our previous findings in autosomal dominant distal hereditary motor neuropathy of a dominant-negative frameshift mutation at the C-terminus of choline transporter that was associated with significantly reduced, but not completely abrogated choline transporter function. Together our findings define divergent neuropathological outcomes arising from different classes of choline transporter mutation with distinct disease processes and modes of inheritance. These findings underscore the essential role played by the choline transporter in sustaining acetylcholine neurotransmission at both central and neuromuscular synapses, with important implications for treatment and drug selection.


Subject(s)
Brain/pathology , Mutation, Missense , Myasthenic Syndromes, Congenital/genetics , Neurodevelopmental Disorders/genetics , Symporters/genetics , Animals , Animals, Genetically Modified , Atrophy , Axons/metabolism , Caenorhabditis elegans , Caenorhabditis elegans Proteins/genetics , Child, Preschool , Female , HEK293 Cells , Homozygote , Humans , Infant , Male , Membrane Transport Proteins/genetics , Pedigree , Presynaptic Terminals/metabolism , Protein Transport , Symporters/metabolism
11.
Brain ; 140(3): 547-554, 2017 03 01.
Article in English | MEDLINE | ID: mdl-28052917

ABSTRACT

Mutations in genes involved in lipid metabolism have increasingly been associated with various subtypes of hereditary spastic paraplegia, a highly heterogeneous group of neurodegenerative motor neuron disorders characterized by spastic paraparesis. Here, we report an unusual autosomal recessive neurodegenerative condition, best classified as a complicated form of hereditary spastic paraplegia, associated with mutation in the ethanolaminephosphotransferase 1 (EPT1) gene (now known as SELENOI), responsible for the final step in Kennedy pathway forming phosphatidylethanolamine from CDP-ethanolamine. Phosphatidylethanolamine is a glycerophospholipid that, together with phosphatidylcholine, constitutes more than half of the total phospholipids in eukaryotic cell membranes. We determined that the mutation defined dramatically reduces the enzymatic activity of EPT1, thereby hindering the final step in phosphatidylethanolamine synthesis. Additionally, due to central nervous system inaccessibility we undertook quantification of phosphatidylethanolamine levels and species in patient and control blood samples as an indication of liver phosphatidylethanolamine biosynthesis. Although this revealed alteration to levels of specific phosphatidylethanolamine fatty acyl species in patients, overall phosphatidylethanolamine levels were broadly unaffected indicating that in blood EPT1 inactivity may be compensated for, in part, via alternate biochemical pathways. These studies define the first human disorder arising due to defective CDP-ethanolamine biosynthesis and provide new insight into the role of Kennedy pathway components in human neurological function.


Subject(s)
Ethanolaminephosphotransferase/genetics , Ethanolaminephosphotransferase/metabolism , Mutation/genetics , Phospholipids/biosynthesis , Signal Transduction/genetics , Spastic Paraplegia, Hereditary/genetics , Adolescent , Child , Child, Preschool , Chromatography, Liquid , Consanguinity , DNA Mutational Analysis , Family Health , Female , Gene Expression , Humans , Infant , Male , Mass Spectrometry , Oman , Phospholipids/blood , Saccharomyces cerevisiae , Spastic Paraplegia, Hereditary/diagnostic imaging , Spastic Paraplegia, Hereditary/enzymology , Spastic Paraplegia, Hereditary/pathology
12.
Brain ; 140(4): 940-952, 2017 Apr 01.
Article in English | MEDLINE | ID: mdl-28334956

ABSTRACT

PRUNE is a member of the DHH (Asp-His-His) phosphoesterase protein superfamily of molecules important for cell motility, and implicated in cancer progression. Here we investigated multiple families from Oman, India, Iran and Italy with individuals affected by a new autosomal recessive neurodevelopmental and degenerative disorder in which the cardinal features include primary microcephaly and profound global developmental delay. Our genetic studies identified biallelic mutations of PRUNE1 as responsible. Our functional assays of disease-associated variant alleles revealed impaired microtubule polymerization, as well as cell migration and proliferation properties, of mutant PRUNE. Additionally, our studies also highlight a potential new role for PRUNE during microtubule polymerization, which is essential for the cytoskeletal rearrangements that occur during cellular division and proliferation. Together these studies define PRUNE as a molecule fundamental for normal human cortical development and define cellular and clinical consequences associated with PRUNE mutation.


Subject(s)
Brain/growth & development , Carrier Proteins/genetics , Developmental Disabilities/genetics , Microcephaly/genetics , Adolescent , Cell Differentiation/genetics , Cell Movement/genetics , Cerebral Cortex/growth & development , Child , Child, Preschool , Cytoskeleton/genetics , Cytoskeleton/ultrastructure , Female , Genes, Recessive , Heredodegenerative Disorders, Nervous System/genetics , Humans , Infant , Male , Microtubules/genetics , Microtubules/ultrastructure , Mutation/genetics , Pedigree , Phosphoric Monoester Hydrolases , Young Adult
13.
Am J Hum Genet ; 94(1): 87-94, 2014 Jan 02.
Article in English | MEDLINE | ID: mdl-24239382

ABSTRACT

The proper development of neuronal circuits during neuromorphogenesis and neuronal-network formation is critically dependent on a coordinated and intricate series of molecular and cellular cues and responses. Although the cortical actin cytoskeleton is known to play a key role in neuromorphogenesis, relatively little is known about the specific molecules important for this process. Using linkage analysis and whole-exome sequencing on samples from families from the Amish community of Ohio, we have demonstrated that mutations in KPTN, encoding kaptin, cause a syndrome typified by macrocephaly, neurodevelopmental delay, and seizures. Our immunofluorescence analyses in primary neuronal cell cultures showed that endogenous and GFP-tagged kaptin associates with dynamic actin cytoskeletal structures and that this association is lost upon introduction of the identified mutations. Taken together, our studies have identified kaptin alterations responsible for macrocephaly and neurodevelopmental delay and define kaptin as a molecule crucial for normal human neuromorphogenesis.


Subject(s)
Developmental Disabilities/genetics , Megalencephaly/genetics , Microfilament Proteins/genetics , Mutation , Seizures/genetics , Actin Cytoskeleton/metabolism , Amino Acid Sequence , Female , Fluorescent Antibody Technique , Genetic Linkage , Humans , Male , Microfilament Proteins/metabolism , Molecular Sequence Data , Pedigree
14.
Pediatr Res ; 81(4): 632-638, 2017 Apr.
Article in English | MEDLINE | ID: mdl-27855150

ABSTRACT

BACKGROUND: The aim was to identify susceptibility alleles for infantile hypertrophic pyloric stenosis (IHPS) in a pedigree previously linked to IHPS5 on chromosome 16q24. METHODS: We screened the positional and functional candidate gene FOXF1 by Sanger sequencing in a single affected individual. All family members for whom DNA was available were genotyped to determine cosegregation status of the putative causal variant. Immunofluorescence studies were performed to compare the cellular localization of wildtype and mutant form of the protein. Transcriptional activity was compared using a luciferase assay. RESULTS: A single novel substitution in FOXF1 (c.416G>A) predicted to result in a missense mutation (R139Q) was shown to cosegregate with disease trait. It was not seen in 560 control chromosomes nor has it been reported in ExAC or ESP. The R139Q substitution affects a conserved arginine residue within the DNA-binding domain of FOXF1. The transcriptional activity of the mutant FOXF1 protein is significantly reduced in comparison to wild-type. CONCLUSION: These results provide strong evidence that the R139Q substitution in FOXF1 causes IHPS in this family and imply a novel pathological pathway for the condition. They further support a role for FOXF1 in the regulation of embryonic and neonatal development of the gastro-intestinal tract.


Subject(s)
Chromosomes, Human, Pair 16 , Forkhead Transcription Factors/genetics , Mutation, Missense , Pyloric Stenosis, Hypertrophic/genetics , Alleles , Arginine/chemistry , Chromosome Mapping , Female , Genetic Variation , Genotype , HEK293 Cells , Haplotypes , Hep G2 Cells , Humans , Male , Microscopy, Fluorescence , Pedigree , Pyloric Stenosis, Hypertrophic/metabolism , Sequence Analysis, DNA , Transcriptional Activation
15.
Hum Mutat ; 37(11): 1157-1161, 2016 11.
Article in English | MEDLINE | ID: mdl-27492651

ABSTRACT

Hereditary spastic paraplegias (HSPs) are genetically and clinically heterogeneous axonopathies primarily affecting upper motor neurons and, in complex forms, additional neurons. Here, we report two families with distinct recessive mutations in TFG, previously suggested to cause HSP based on findings in a single small family with complex HSP. The first carried a homozygous c.317G>A (p.R106H) variant and presented with pure HSP. The second carried the same homozygous c.316C>T (p.R106C) variant previously reported and displayed a similarly complex phenotype including optic atrophy. Haplotyping and bisulfate sequencing revealed evidence for a c.316C>T founder allele, as well as for a c.316_317 mutation hotspot. Expression of mutant TFG proteins in cultured neurons revealed mitochondrial fragmentation, the extent of which correlated with clinical severity. Our findings confirm the causal nature of bi-allelic TFG mutations for HSP, broaden the clinical and mutational spectra, and suggest mitochondrial impairment to represent a pathomechanistic link to other neurodegenerative conditions.


Subject(s)
Mutation, Missense , Proteins/genetics , Proteins/metabolism , Spastic Paraplegia, Hereditary/pathology , Animals , Cells, Cultured , Female , Genetic Predisposition to Disease , Humans , Magnetic Resonance Imaging/methods , Male , Mice , Mitochondria/pathology , Neurons/cytology , Neurons/metabolism , Neurons/pathology , Pedigree , Sequence Analysis, DNA , Spastic Paraplegia, Hereditary/genetics , Spastic Paraplegia, Hereditary/metabolism
16.
BMC Med Genet ; 17(1): 82, 2016 Nov 16.
Article in English | MEDLINE | ID: mdl-27852232

ABSTRACT

BACKGROUND: CMT-2 is a clinically and genetically heterogeneous group of peripheral axonal neuropathies characterized by slowly progressive weakness and atrophy of distal limb muscles resulting from length-dependent motor and sensory neurodegeneration. Classical giant axonal neuropathy (GAN) is an autosomal recessively inherited progressive neurodegenerative disorder of the peripheral and central nervous systems, typically diagnosed in early childhood and resulting in death by the end of the third decade. Distinctive phenotypic features are the presence of "kinky" hair and long eyelashes. The genetic basis of the disease has been well established, with over 40 associated mutations identified in the gene GAN, encoding the BTB-KELCH protein gigaxonin, involved in intermediate filament regulation. METHODS: An Illumina Human CytoSNP-12 array followed by whole exome sequence analysis was used to identify the disease associated gene mutation in a large consanguineous family diagnosed with Charcot-Marie-Tooth disease type 2 (CMT-2) from which all but one affected member had straight hair. RESULTS: Here we report the identification of a novel GAN missense mutation underlying the CMT-2 phenotype observed in this family. Although milder forms of GAN, with and without the presence of kinky hair have been reported previously, a phenotype distinct from that was investigated in this study. All family members lacked common features of GAN, including ataxia, nystagmus, intellectual disability, seizures, and central nervous system involvement. CONCLUSIONS: Our findings broaden the spectrum of phenotypes associated with GAN mutations and emphasize a need to proceed with caution when providing families with diagnostic or prognostic information based on either clinical or genetic findings alone.


Subject(s)
Charcot-Marie-Tooth Disease/genetics , Consanguinity , Cytoskeletal Proteins/genetics , Mutation, Missense/genetics , Adult , Alleles , Animals , Brain/diagnostic imaging , Charcot-Marie-Tooth Disease/pathology , Child , Child, Preschool , Electrophysiological Phenomena , Genotype , Humans , Israel , Male , Molecular Sequence Data , Pedigree , Phenotype , Polymorphism, Single Nucleotide , Sequence Alignment , Twins, Dizygotic
17.
Brain ; 138(Pt 8): 2173-90, 2015 Aug.
Article in English | MEDLINE | ID: mdl-26070982

ABSTRACT

We describe a novel nephrocerebellar syndrome on the Galloway-Mowat syndrome spectrum among 30 children (ages 1.0 to 28 years) from diverse Amish demes. Children with nephrocerebellar syndrome had progressive microcephaly, visual impairment, stagnant psychomotor development, abnormal extrapyramidal movements and nephrosis. Fourteen died between ages 2.7 and 28 years, typically from renal failure. Post-mortem studies revealed (i) micrencephaly without polymicrogyria or heterotopia; (ii) atrophic cerebellar hemispheres with stunted folia, profound granule cell depletion, Bergmann gliosis, and signs of Purkinje cell deafferentation; (iii) selective striatal cholinergic interneuron loss; and (iv) optic atrophy with delamination of the lateral geniculate nuclei. Renal tissue showed focal and segmental glomerulosclerosis and extensive effacement and microvillus transformation of podocyte foot processes. Nephrocerebellar syndrome mapped to 700 kb on chromosome 15, which contained a single novel homozygous frameshift variant (WDR73 c.888delT; p.Phe296Leufs*26). WDR73 protein is expressed in human cerebral cortex, hippocampus, and cultured embryonic kidney cells. It is concentrated at mitotic microtubules and interacts with α-, ß-, and γ-tubulin, heat shock proteins 70 and 90 (HSP-70; HSP-90), and the carbamoyl phosphate synthetase 2/aspartate transcarbamylase/dihydroorotase multi-enzyme complex. Recombinant WDR73 p.Phe296Leufs*26 and p.Arg256Profs*18 proteins are truncated, unstable, and show increased interaction with α- and ß-tubulin and HSP-70/HSP-90. Fibroblasts from patients homozygous for WDR73 p.Phe296Leufs*26 proliferate poorly in primary culture and senesce early. Our data suggest that in humans, WDR73 interacts with mitotic microtubules to regulate cell cycle progression, proliferation and survival in brain and kidney. We extend the Galloway-Mowat syndrome spectrum with the first description of diencephalic and striatal neuropathology.


Subject(s)
Brain/metabolism , Cell Cycle/genetics , Hernia, Hiatal/genetics , Microcephaly/genetics , Mutation/genetics , Nephrosis/genetics , Proteins/metabolism , Adolescent , Adult , Child , Child, Preschool , Female , Glomerulosclerosis, Focal Segmental/genetics , Glomerulosclerosis, Focal Segmental/metabolism , Homozygote , Humans , Infant , Male , Proteins/genetics , Tubulin/genetics , Young Adult
18.
BMC Med Genet ; 16: 104, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26554554

ABSTRACT

BACKGROUND: The deletion of the chromosome 4p16.3 Wolf-Hirschhorn syndrome critical region (WHSCR-2) typically results in a characteristic facial appearance, varying intellectual disability, stereotypies and prenatal onset of growth retardation, while gains of the same chromosomal region result in a more variable degree of intellectual deficit and dysmorphism. Similarly the phenotype of individuals with terminal deletions of distal chromosome 3p (3p deletion syndrome) varies from mild to severe intellectual deficit, micro- and trigonocephaly, and a distinct facial appearance. METHODS AND RESULTS: We investigated a large Indian five-generation pedigree with ten affected family members in which chromosomal microarray and fluorescence in situ hybridization analyses disclosed a complex rearrangement involving chromosomal subregions 4p16.1 and 3p26.3 resulting in a 4p16.1 deletion and 3p26.3 microduplication in three individuals, and a 4p16.1 duplication and 3p26.3 microdeletion in seven individuals. A typical clinical presentation of WHS was observed in all three cases with 4p16.1 deletion and 3p26.3 microduplication. Individuals with a 4p16.1 duplication and 3p26.3 microdeletion demonstrated a range of clinical features including typical 3p microdeletion or 4p partial trisomy syndrome to more severe neurodevelopmental delay with distinct dysmorphic features. CONCLUSION: We present the largest pedigree with complex t(4p;3p) chromosomal rearrangements and diverse clinical outcomes including Wolf Hirschorn-, 3p deletion-, and 4p duplication syndrome amongst affected individuals.


Subject(s)
Chromosome Disorders/genetics , Chromosomes, Human, Pair 3 , Chromosomes, Human, Pair 4 , Chromosome Disorders/etiology , Developmental Disabilities/genetics , Female , Humans , In Situ Hybridization, Fluorescence , India , Male , Oligonucleotide Array Sequence Analysis , Pedigree , Sequence Deletion , Trisomy , Wolf-Hirschhorn Syndrome/genetics
19.
Am J Med Genet A ; 164A(4): 1003-9, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24458550

ABSTRACT

Germline mutations in the gene CBL (Casitas B-lineage lymphoma), involved in the RAS-MAPK signaling pathway, have been found as a rare cause of the neuro-cardio-facial-cutaneous syndromes. Somatically acquired homozygous CBL mutations were initially identified in association with myeloproliferative disorders, particularly juvenile myelomonocytic leukemia (JMML). We describe a girl with a Noonan-like phenotype of bilateral ptosis, lymphedema of the lower limbs and moderate intellectual disability, due to a de novo heterozygous mutation in CBL. She developed an ovarian mixed germ cell/teratoma with later occurrence of mature liver, omental, and ovarian teratomas. Copy neutral loss of heterozygosity for the CBL mutation due to acquired segmental uniparental disomy of 11q23 was observed in three teratomas, suggesting a specific association of CBL mutations in germ cell tumor predisposition.


Subject(s)
Chromosomes, Human, Pair 11 , Germ-Line Mutation , Lymphedema/genetics , Ovarian Neoplasms/genetics , Proto-Oncogene Proteins c-cbl/genetics , Teratoma/genetics , Uniparental Disomy/genetics , Adolescent , Female , Heterozygote , Humans
20.
J Med Genet ; 50(2): 65-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-23243086

ABSTRACT

BACKGROUND: Deregulation of the activity of the ubiquitin ligase E6AP (UBE3A) is well recognised to contribute to the development of Angelman syndrome (AS). The ubiquitin ligase HERC2, encoded by the HERC2 gene is thought to be a key regulator of E6AP. METHODS AND RESULTS: Using a combination of autozygosity mapping and linkage analysis, we studied an autosomal-recessive neurodevelopmental disorder with some phenotypic similarities to AS, found among the Old Order Amish. Our molecular investigation identified a mutation in HERC2 associated with the disease phenotype. We establish that the encoded mutant HERC2 protein has a reduced half-life compared with its wild-type counterpart, which is associated with a significant reduction in HERC2 levels in affected individuals. CONCLUSIONS: Our data implicate a model in which disruption of HERC2 function relates to a reduction in E6AP activity resulting in neurodevelopmental delay, suggesting a previously unrecognised role of HERC2 in the pathogenesis of AS.


Subject(s)
Amish/genetics , Angelman Syndrome/genetics , Guanine Nucleotide Exchange Factors/genetics , Mutation , Adolescent , Adult , Cell Cycle Proteins/chemistry , Cell Line , Child , Child, Preschool , DNA/analysis , DNA/genetics , DNA Mutational Analysis , Female , Fibroblasts/chemistry , Fibroblasts/metabolism , Genome-Wide Association Study , Guanine Nucleotide Exchange Factors/blood , Guanine Nucleotide Exchange Factors/chemistry , Guanine Nucleotide Exchange Factors/metabolism , HEK293 Cells , Humans , Infant , Male , Models, Molecular , Nuclear Proteins/chemistry , Pedigree , Ubiquitin-Protein Ligases
SELECTION OF CITATIONS
SEARCH DETAIL