Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 36
Filter
2.
Genet Med ; 25(10): 100927, 2023 10.
Article in English | MEDLINE | ID: mdl-37422718

ABSTRACT

PURPOSE: The SF3B splicing complex is composed of SF3B1-6 and PHF5A. We report a developmental disorder caused by de novo variants in PHF5A. METHODS: Clinical, genomic, and functional studies using subject-derived fibroblasts and a heterologous cellular system were performed. RESULTS: We studied 9 subjects with congenital malformations, including preauricular tags and hypospadias, growth abnormalities, and developmental delay who had de novo heterozygous PHF5A variants, including 4 loss-of-function (LOF), 3 missense, 1 splice, and 1 start-loss variant. In subject-derived fibroblasts with PHF5A LOF variants, wild-type and variant PHF5A mRNAs had a 1:1 ratio, and PHF5A mRNA levels were normal. Transcriptome sequencing revealed alternative promoter use and downregulated genes involved in cell-cycle regulation. Subject and control fibroblasts had similar amounts of PHF5A with the predicted wild-type molecular weight and of SF3B1-3 and SF3B6. SF3B complex formation was unaffected in 2 subject cell lines. CONCLUSION: Our data suggest the existence of feedback mechanisms in fibroblasts with PHF5A LOF variants to maintain normal levels of SF3B components. These compensatory mechanisms in subject fibroblasts with PHF5A or SF3B4 LOF variants suggest disturbed autoregulation of mutated splicing factor genes in specific cell types, that is, neural crest cells, during embryonic development rather than haploinsufficiency as pathomechanism.


Subject(s)
Craniofacial Abnormalities , Hypospadias , Male , Humans , Hypospadias/genetics , RNA Splicing Factors/genetics , RNA Splicing , Transcription Factors/genetics , Transcription Factors/metabolism , Trans-Activators/genetics , RNA-Binding Proteins/genetics
3.
ChemistryOpen ; 10(9): 889-895, 2021 09.
Article in English | MEDLINE | ID: mdl-34468091

ABSTRACT

Due to their special chemical structure, tetraether lipids (TEL) represent essential elements of archaeal membranes, providing these organisms with extraordinary properties. Here we describe the characterization of a newly isolated structural element of the main lipids. The TEL fragment GDNT-ß-Glu was isolated from Sulfolobus metallicus and characterized in terms of its chemical structure by NMR- and MS-investigations. The obtained data are dissimilar to analogically derived established structures - in essence, the binding relationships in the polar head group are re-determined and verified. With this work, we provide an important contribution to the structure elucidation of intact TEL also contained in other Sulfolobus strains such as Solfulobus acidocaldarius and Sulfolobus solfataricus.


Subject(s)
Diglycerides/chemistry , Glycolipids/chemistry , Membrane Lipids/chemistry , Sulfolobus/chemistry , Carbon-13 Magnetic Resonance Spectroscopy , Cyclization , Diglycerides/isolation & purification , Glycolipids/isolation & purification , Mass Spectrometry , Membrane Lipids/isolation & purification , Sulfolobus/classification
4.
J Inherit Metab Dis ; 44(4): 972-986, 2021 07.
Article in English | MEDLINE | ID: mdl-33320377

ABSTRACT

Several inborn errors of metabolism show cutis laxa as a highly recognizable feature. One group of these metabolic cutis laxa conditions is autosomal recessive cutis laxa type 2 caused by defects in v-ATPase components or the mitochondrial proline cycle. Besides cutis laxa, muscular hypotonia and cardiac abnormalities are hallmarks of autosomal recessive cutis laxa type 2D (ARCL2D) due to pathogenic variants in ATP6V1A encoding subunit A of the v-ATPase. Here, we report on three affected individuals from two families with ARCL2D in whom we performed whole exome and Sanger sequencing. We performed functional studies in fibroblasts from one individual, summarized all known probands' clinical, molecular, and biochemical features and compared them, also to other metabolic forms of cutis laxa. We identified novel missense and the first nonsense variant strongly affecting ATP6V1A expression. All six ARCL2D affected individuals show equally severe cutis laxa and dysmorphism at birth. While for one no information was available, two died in infancy and three are now adolescents with mild or absent intellectual disability. Muscular weakness, ptosis, contractures, and elevated muscle enzymes indicated a persistent myopathy. In cellular studies, a fragmented Golgi compartment, a delayed Brefeldin A-induced retrograde transport and glycosylation abnormalities were present in fibroblasts from two individuals. This is the second and confirmatory report on pathogenic variants in ATP6V1A as the cause of this extremely rare condition and the first to describe a nonsense allele. Our data highlight the tremendous clinical variability of ATP6V1A related phenotypes even within the same family.


Subject(s)
Cutis Laxa/genetics , Mutation, Missense , Vacuolar Proton-Translocating ATPases/genetics , Adolescent , Alleles , Case-Control Studies , Fibroblasts/metabolism , Golgi Apparatus/metabolism , Humans , Infant , Infant, Newborn , Intellectual Disability/genetics , Male , Pedigree , Phenotype
5.
Neurodegener Dis ; 20(1): 35-38, 2020.
Article in English | MEDLINE | ID: mdl-32623435

ABSTRACT

INTRODUCTION: Familial isolated deficiency of vitamin E (VED or AVED; MIM #277460) is a progressive neurodegenerative disorder resembling Friedreich ataxia. It is caused by the deficiency of α-tocopherol transfer protein that prevents patients from retaining vitamin E. Oral vitamin E supplements are an accepted treatment, but detailed dosage recommendations and reports on long-term therapeutic results are scarce. METHODS: The first patient with VED was discovered at our institution at the age of 12 years and has since been followed with clinical, neurophysiological, neuroradiological, and biochemical investigations to his present age of 52 years. For the last 36 years, the patient has scrupulously followed a custom-made high-dose vitamin E supplement regimen that we devised on the basis of studies of his metabolism of vitamin E. RESULTS: Over the long period of observation, the patient has remained in good general health and has not shown progression of neurological symptoms and signs. His vitamin E plasma levels were always moderately above the normal range. During short interruptions of vitamin E supplements, vitamin E levels fell rapidly, even after years of massive supplementation. DISCUSSION: In this VED patient, a specified and carefully controlled high-dose vitamin E therapy has prevented any recognizable progression of the neurodegenerative process over more than 3 decades of observation.


Subject(s)
Ataxia/drug therapy , Ataxia/genetics , Vitamin E Deficiency/drug therapy , Vitamin E Deficiency/genetics , Vitamin E/therapeutic use , Adolescent , Adult , Child , Dietary Supplements , Humans , Male , Middle Aged , Time Factors , Young Adult
6.
PLoS Genet ; 13(4): e1006746, 2017 04.
Article in English | MEDLINE | ID: mdl-28453519

ABSTRACT

Mid-hindbrain malformations can occur during embryogenesis through a disturbance of transient and localized gene expression patterns within these distinct brain structures. Rho guanine nucleotide exchange factor (ARHGEF) family members are key for controlling the spatiotemporal activation of Rho GTPase, to modulate cytoskeleton dynamics, cell division, and cell migration. We identified, by means of whole exome sequencing, a homozygous frameshift mutation in the ARHGEF2 as a cause of intellectual disability, a midbrain-hindbrain malformation, and mild microcephaly in a consanguineous pedigree of Kurdish-Turkish descent. We show that loss of ARHGEF2 perturbs progenitor cell differentiation and that this is associated with a shift of mitotic spindle plane orientation, putatively favoring more symmetric divisions. The ARHGEF2 mutation leads to reduction in the activation of the RhoA/ROCK/MLC pathway crucial for cell migration. We demonstrate that the human brain malformation is recapitulated in Arhgef2 mutant mice and identify an aberrant migration of distinct components of the precerebellar system as a pathomechanism underlying the midbrain-hindbrain phenotype. Our results highlight the crucial function of ARHGEF2 in human brain development and identify a mutation in ARHGEF2 as novel cause of a neurodevelopmental disorder.


Subject(s)
Cell Movement/genetics , Frameshift Mutation/genetics , Intellectual Disability/genetics , Rho Guanine Nucleotide Exchange Factors/genetics , Animals , Cytoskeleton/genetics , Exome/genetics , Female , High-Throughput Nucleotide Sequencing , Homozygote , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/pathology , Magnetic Resonance Imaging , Male , Mesencephalon/diagnostic imaging , Mesencephalon/pathology , Mice , Pedigree , Rhombencephalon/diagnostic imaging , Rhombencephalon/pathology , Signal Transduction , rhoA GTP-Binding Protein/genetics
7.
Eur J Hum Genet ; 25(2): 253-256, 2017 02.
Article in English | MEDLINE | ID: mdl-27827379

ABSTRACT

Klüver-Bucy syndrome (KBS) comprises a set of neurobehavioral symptoms with psychic blindness, hypersexuality, disinhibition, hyperorality, and hypermetamorphosis that were originally observed after bilateral lobectomy in Rhesus monkeys. We investigated two siblings with KBS from a consanguineous family by whole-exome sequencing and autozygosity mapping. We detected a homozygous variant in the heparan-α-glucosaminidase-N-acetyltransferase gene (HGSNAT; c.518G>A, p.(G173D), NCBI ClinVar RCV000239404.1), which segregated with the phenotype. Disease-causing variants in this gene are known to be associated with autosomal recessive Mucopolysaccharidosis type IIIC (MPSIIIC, Sanfilippo C). This lysosomal storage disease is due to deficiency of the acetyl-CoA:α-glucosaminidase-N-acetyltransferase, which was shown to be reduced in patient fibroblasts. Our report extends the phenotype associated with MPSIIIC. Besides MPSIIIA and MPSIIIB, due to variants in SGSH and NAGLU, this is the third subtype of Sanfilippo disease to be associated with KBS. MPSIII should be included in the differential diagnosis of young patients with KBS.


Subject(s)
Acetyltransferases/genetics , Kluver-Bucy Syndrome/genetics , Mucopolysaccharidosis III/genetics , Child , Exome , Female , Genes, Recessive , Homozygote , Humans , Kluver-Bucy Syndrome/complications , Kluver-Bucy Syndrome/diagnosis , Male , Mucopolysaccharidosis III/complications , Mucopolysaccharidosis III/diagnosis , Phenotype , Siblings
8.
Elife ; 52016 08 06.
Article in English | MEDLINE | ID: mdl-27495975

ABSTRACT

Mitochondriopathies often present clinically as multisystemic disorders of primarily high-energy consuming organs. Assembly, turnover, and surveillance of mitochondrial proteins are essential for mitochondrial function and a key task of AAA family members of metalloproteases. We identified a homozygous mutation in the nuclear encoded mitochondrial escape 1-like 1 gene YME1L1, member of the AAA protease family, as a cause of a novel mitochondriopathy in a consanguineous pedigree of Saudi Arabian descent. The homozygous missense mutation, located in a highly conserved region in the mitochondrial pre-sequence, inhibits cleavage of YME1L1 by the mitochondrial processing peptidase, which culminates in the rapid degradation of YME1L1 precursor protein. Impaired YME1L1 function causes a proliferation defect and mitochondrial network fragmentation due to abnormal processing of OPA1. Our results identify mutations in YME1L1 as a cause of a mitochondriopathy with optic nerve atrophy highlighting the importance of YME1L1 for mitochondrial functionality in humans.


Subject(s)
Homozygote , Metalloendopeptidases/genetics , Mitochondria/pathology , Mitochondrial Diseases/genetics , Mutation, Missense , Optic Atrophy/genetics , ATPases Associated with Diverse Cellular Activities , Female , Humans , Male , Mitochondrial Proteins
9.
Am J Hum Genet ; 98(3): 473-489, 2016 Mar 03.
Article in English | MEDLINE | ID: mdl-26924529

ABSTRACT

Transcriptional signal cointegrators associate with transcription factors or nuclear receptors and coregulate tissue-specific gene transcription. We report on recessive loss-of-function mutations in two genes (TRIP4 and ASCC1) that encode subunits of the nuclear activating signal cointegrator 1 (ASC-1) complex. We used autozygosity mapping and whole-exome sequencing to search for pathogenic mutations in four families. Affected individuals presented with prenatal-onset spinal muscular atrophy (SMA), multiple congenital contractures (arthrogryposis multiplex congenita), respiratory distress, and congenital bone fractures. We identified homozygous and compound-heterozygous nonsense and frameshift TRIP4 and ASCC1 mutations that led to a truncation or the entire absence of the respective proteins and cosegregated with the disease phenotype. Trip4 and Ascc1 have identical expression patterns in 17.5-day-old mouse embryos with high expression levels in the spinal cord, brain, paraspinal ganglia, thyroid, and submandibular glands. Antisense morpholino-mediated knockdown of either trip4 or ascc1 in zebrafish disrupted the highly patterned and coordinated process of α-motoneuron outgrowth and formation of myotomes and neuromuscular junctions and led to a swimming defect in the larvae. Immunoprecipitation of the ASC-1 complex consistently copurified cysteine and glycine rich protein 1 (CSRP1), a transcriptional cofactor, which is known to be involved in spinal cord regeneration upon injury in adult zebrafish. ASCC1 mutant fibroblasts downregulated genes associated with neurogenesis, neuronal migration, and pathfinding (SERPINF1, DAB1, SEMA3D, SEMA3A), as well as with bone development (TNFRSF11B, RASSF2, STC1). Our findings indicate that the dysfunction of a transcriptional coactivator complex can result in a clinical syndrome affecting the neuromuscular system.


Subject(s)
Fractures, Bone/genetics , Gene Expression Regulation, Developmental , Muscular Atrophy, Spinal/genetics , Transcription Factors/genetics , Amino Acid Sequence , Animals , Arthrogryposis/diagnosis , Arthrogryposis/genetics , Carrier Proteins/genetics , Cells, Cultured , Fibroblasts/cytology , Fibroblasts/metabolism , Fractures, Bone/diagnosis , Gene Expression Profiling , Homozygote , Humans , LIM Domain Proteins/genetics , Mice , Molecular Sequence Data , Muscular Atrophy, Spinal/diagnosis , Mutation , Nuclear Proteins/genetics , Pedigree , Phenotype , Zebrafish , Zebrafish Proteins/genetics
10.
Eur J Hum Genet ; 23(10): 1308-17, 2015 Oct.
Article in English | MEDLINE | ID: mdl-25758992

ABSTRACT

Congenital cardiac and neurodevelopmental deficits have been recently linked to the mediator complex subunit 13-like protein MED13L, a subunit of the CDK8-associated mediator complex that functions in transcriptional regulation through DNA-binding transcription factors and RNA polymerase II. Heterozygous MED13L variants cause transposition of the great arteries and intellectual disability (ID). Here, we report eight patients with predominantly novel MED13L variants who lack such complex congenital heart malformations. Rather, they depict a syndromic form of ID characterized by facial dysmorphism, ID, speech impairment, motor developmental delay with muscular hypotonia and behavioral difficulties. We thereby define a novel syndrome and significantly broaden the clinical spectrum associated with MED13L variants. A prominent feature of the MED13L neurocognitive presentation is profound language impairment, often in combination with articulatory deficits.


Subject(s)
Abnormalities, Multiple/genetics , Mediator Complex/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Intellectual Disability/genetics , Male , Muscle Hypotonia/genetics , Mutation/genetics , Phenotype , Syndrome , Transposition of Great Vessels/genetics
11.
Orphanet J Rare Dis ; 9: 113, 2014 Oct 21.
Article in English | MEDLINE | ID: mdl-25332050

ABSTRACT

Warburg micro syndrome (WARBM) is a genetic heterogeneous disease characterized by microcephaly, intellectual disability, brain, ocular, and endocrine anomalies. WARBM1-4 can be caused by biallelic mutations of the RAB3GAP1 (RAB3 GTPase-activating protein 1), RAB3GAP2, RAB18 (RAS-associated protein RAB18), or TBC1D20 (TBC1 domain protein, member 20) gene, respectively. Here, we delineate the so far largest intragenic homozygous RAB3GAP1 microdeletion. Despite the size of the RAB3GAP1 gene deletion, the patient phenotype is mainly consistent with that of other WARBM1 patients, supporting strongly the theory that WARBM1 is caused by a loss of RAB3GAP1 function. We further highlight osteopenia as a feature of WARBM1.


Subject(s)
Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/genetics , Cataract/congenital , Cornea/abnormalities , Gene Deletion , Homozygote , Hypogonadism/diagnosis , Hypogonadism/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics , Microcephaly/diagnosis , Microcephaly/genetics , Optic Atrophy/diagnosis , Optic Atrophy/genetics , rab3 GTP-Binding Proteins/genetics , Cataract/diagnosis , Cataract/genetics , Child , Child, Preschool , Female , Humans , Male , Pedigree
12.
J Med Genet ; 51(6): 375-87, 2014 Jun.
Article in English | MEDLINE | ID: mdl-24714694

ABSTRACT

BACKGROUND: NKX2-1 encodes a transcription factor with large impact on the development of brain, lung and thyroid. Germline mutations of NKX2-1 can lead to dysfunction and malformations of these organs. Starting from the largest coherent collection of patients with a suspected phenotype to date, we systematically evaluated frequency, quality and spectrum of phenotypic consequences of NKX2-1 mutations. METHODS: After identifying mutations by Sanger sequencing and array CGH, we comprehensively reanalysed the phenotype of affected patients and their relatives. We employed electrophoretic mobility shift assay (EMSA) to detect alterations of NKX2-1 DNA binding. Gene expression was monitored by means of in situ hybridisation and compared with the expression level of MBIP, a candidate gene presumably involved in the disorders and closely located in close genomic proximity to NKX2-1. RESULTS: Within 101 index patients, we detected 17 point mutations and 10 deletions. Neurological symptoms were the most consistent finding (100%), followed by lung affection (78%) and thyroidal dysfunction (75%). Novel symptoms associated with NKX2-1 mutations comprise abnormal height, bouts of fever and cardiac septum defects. In contrast to previous reports, our data suggest that missense mutations in the homeodomain of NKX2-1 not necessarily modify its DNA binding capacity and that this specific type of mutations may be associated with mild pulmonary phenotypes such as asthma. Two deletions did not include NKX2-1, but MBIP, whose expression spatially and temporarily coincides with NKX2-1 in early murine development. CONCLUSIONS: The high incidence of NKX2-1 mutations strongly recommends the routine screen for mutations in patients with corresponding symptoms. However, this analysis should not be confined to the exonic sequence alone, but should take advantage of affordable NGS technology to expand the target to adjacent regulatory sequences and the NKX2-1 interactome in order to maximise the yield of this diagnostic effort.


Subject(s)
Genetic Diseases, Inborn , Nuclear Proteins/genetics , Transcription Factors/genetics , Adolescent , Child , Child, Preschool , Comparative Genomic Hybridization , DNA Copy Number Variations/genetics , Electrophoretic Mobility Shift Assay , Female , Gene Deletion , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/physiopathology , Humans , Infant , Infant, Newborn , Male , Phenotype , Point Mutation/genetics , Thyroid Nuclear Factor 1
14.
J Med Genet ; 51(4): 275-82, 2014 Apr.
Article in English | MEDLINE | ID: mdl-24556084

ABSTRACT

BACKGROUND: Congenital muscular dystrophies (CMD) with hypoglycosylation of α-dystroglycan are clinically and genetically heterogeneous disorders that are often associated with brain malformations and eye defects. Presently, 16 proteins are known whose dysfunction impedes glycosylation of α-dystroglycan and leads to secondary dystroglycanopathy. OBJECTIVE: To identify the cause of CMD with secondary merosin deficiency, hypomyelination and intellectual disability in two siblings from a consanguineous family. METHODS: Autozygosity mapping followed by whole exome sequencing and immunochemistry were used to discover and verify a new genetic defect in two siblings with CMD. RESULTS: We identified a homozygous missense mutation (c.325C>T, p.Q109*) in protein O-mannosyl kinase (POMK) that encodes a glycosylation-specific kinase (SGK196) required for function of the dystroglycan complex. The protein was absent from skeletal muscle and skin fibroblasts of the patients. In patient muscle, ß-dystroglycan was normally expressed at the sarcolemma, while α-dystroglycan failed to do so. Further, we detected co-localisation of POMK with desmin at the costameres in healthy muscle, and a substantial loss of desmin from the patient muscle. CONCLUSIONS: Homozygous truncating mutations in POMK lead to CMD with secondary merosin deficiency, hypomyelination and intellectual disability. Loss of desmin suggests that failure of proper α-dystroglycan glycosylation impedes the binding to extracellular matrix proteins and also affects the cytoskeleton.


Subject(s)
Hearing Loss/complications , Intellectual Disability/complications , Laminin/deficiency , Muscular Dystrophies/congenital , Mutation/genetics , Myelin Sheath/pathology , Protein Kinases/genetics , Adolescent , Base Sequence , Child , Child, Preschool , DNA Mutational Analysis , Family , Female , Hearing Loss/enzymology , Hearing Loss/genetics , Humans , Immunohistochemistry , Infant , Infant, Newborn , Intellectual Disability/enzymology , Intellectual Disability/genetics , Male , Molecular Sequence Data , Muscle, Skeletal/pathology , Muscular Dystrophies/complications , Muscular Dystrophies/enzymology , Muscular Dystrophies/genetics , Pedigree , Young Adult
15.
Meta Gene ; 2: 160-3, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25606399

ABSTRACT

•47,XYY syndrome is a frequent sex chromosome aneuploidy.•Overview of characteristic symptoms of 47,XXY•First report of 47,XYY and microcephaly in a preterm child•Brief differential diagnosis of microcephaly.

16.
Ann Clin Transl Neurol ; 1(12): 1024-35, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25574476

ABSTRACT

OBJECTIVE: To identify the cause of a so-far unreported phenotype of infantile-onset multisystem neurologic, endocrine, and pancreatic disease (IMNEPD). METHODS: We characterized a consanguineous family of Yazidian-Turkish descent with IMNEPD. The two affected children suffer from intellectual disability, postnatal microcephaly, growth retardation, progressive ataxia, distal muscle weakness, peripheral demyelinating sensorimotor neuropathy, sensorineural deafness, exocrine pancreas insufficiency, hypothyroidism, and show signs of liver fibrosis. We performed whole-exome sequencing followed by bioinformatic analysis and Sanger sequencing on affected and unaffected family members. The effect of mutations in the candidate gene was studied in wild-type and mutant mice and in patient and control fibroblasts. RESULTS: In a consanguineous family with two individuals with IMNEPD, we identified a homozygous frameshift mutation in the previously not disease-associated peptidyl-tRNA hydrolase 2 (PTRH2) gene. PTRH2 encodes a primarily mitochondrial protein involved in integrin-mediated cell survival and apoptosis signaling. We show that PTRH2 is highly expressed in the developing brain and is a key determinant in maintaining cell survival during human tissue development. Moreover, we link PTRH2 to the mTOR pathway and thus the control of cell size. The pathology suggested by the human phenotype and neuroimaging studies is supported by analysis of mutant mice and patient fibroblasts. INTERPRETATION: We report a novel disease phenotype, show that the genetic cause is a homozygous mutation in the PTRH2 gene, and demonstrate functional effects in mouse and human tissues. Mutations in PTRH2 should be considered in patients with undiagnosed multisystem neurologic, endocrine, and pancreatic disease.

17.
Gene ; 2013 Sep 12.
Article in English | MEDLINE | ID: mdl-24035900

ABSTRACT

This article has been withdrawn at the request of the author(s) and/or editor. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.

18.
Am J Hum Genet ; 92(5): 681-95, 2013 May 02.
Article in English | MEDLINE | ID: mdl-23623388

ABSTRACT

Arthrogryposis multiplex congenita (AMC) is caused by heterogeneous pathologies leading to multiple antenatal joint contractures through fetal akinesia. Understanding the pathophysiology of this disorder is important for clinical care of the affected individuals and genetic counseling of the families. We thus aimed to establish the genetic basis of an AMC subtype that is associated with multiple dysmorphic features and intellectual disability (ID). We used haplotype analysis, next-generation sequencing, array comparative genomic hybridization, and chromosome breakpoint mapping to identify the pathogenic mutations in families and simplex cases. Suspected disease variants were verified by cosegregation analysis. We identified disease-causing mutations in the zinc-finger gene ZC4H2 in four families affected by X-linked AMC plus ID and one family affected by cerebral palsy. Several heterozygous females were also affected, but to a lesser degree. Furthermore, we found two ZC4H2 deletions and one rearrangement in two female and one male unrelated simplex cases, respectively. In mouse primary hippocampal neurons, transiently produced ZC4H2 localized to the postsynaptic compartment of excitatory synapses, and the altered protein influenced dendritic spine density. In zebrafish, antisense-morpholino-mediated zc4h2 knockdown caused abnormal swimming and impaired α-motoneuron development. All missense mutations identified herein failed to rescue the swimming defect of zebrafish morphants. We conclude that ZC4H2 point mutations, rearrangements, and small deletions cause a clinically variable broad-spectrum neurodevelopmental disorder of the central and peripheral nervous systems in both familial and simplex cases of both sexes. Our results highlight the importance of ZC4H2 for genetic testing of individuals presenting with ID plus muscle weakness and minor or major forms of AMC.


Subject(s)
Abnormalities, Multiple/genetics , Arthrogryposis/genetics , Carrier Proteins/genetics , Genetic Predisposition to Disease/genetics , Intellectual Disability/genetics , Neuronal Plasticity/genetics , Zinc Fingers/genetics , Abnormalities, Multiple/pathology , Animals , Arthrogryposis/pathology , Cells, Cultured , Chromosome Breakpoints , Comparative Genomic Hybridization , Female , Haplotypes/genetics , High-Throughput Nucleotide Sequencing , Humans , Immunoblotting , In Situ Hybridization , Intellectual Disability/pathology , Intracellular Signaling Peptides and Proteins , Male , Mice , Mutation/genetics , Nuclear Proteins , Pedigree , Synapses/genetics , Zebrafish
19.
Neurology ; 80(5): 438-46, 2013 Jan 29.
Article in English | MEDLINE | ID: mdl-23284067

ABSTRACT

OBJECTIVES: Pontocerebellar hypoplasia with spinal muscular atrophy, also known as PCH1, is a group of autosomal recessive disorders characterized by generalized muscle weakness and global developmental delay commonly resulting in early death. Gene defects had been discovered only in single patients until the recent identification of EXOSC3 mutations in several families with relatively mild course of PCH1. We aim to genetically stratify subjects in a large and well-defined cohort to define the clinical spectrum and genotype-phenotype correlation. METHODS: We documented clinical, neuroimaging, and morphologic data of 37 subjects from 27 families with PCH1. EXOSC3 gene sequencing was performed in 27 unrelated index patients of mixed ethnicity. RESULTS: Biallelic mutations in EXOSC3 were detected in 10 of 27 families (37%). The most common mutation among all ethnic groups was c.395A>C, p.D132A, responsible for 11 (55%) of the 20 mutated alleles and ancestral in origin. The mutation-positive subjects typically presented with normal pregnancy, normal birth measurements, and relative preservation of brainstem and cortical structures. Psychomotor retardation was profound in all patients but lifespan was variable, with 3 subjects surviving beyond the late teens. Abnormal oculomotor function was commonly observed in patients surviving beyond the first year. Major clinical features previously reported in PCH1, including intrauterine abnormalities, postnatal hypoventilation and feeding difficulties, joint contractures, and neonatal death, were rarely observed in mutation-positive infants but were typical among the mutation-negative subjects. CONCLUSION: EXOSC3 mutations account for 30%-40% of patients with PCH1 with variability in survival and clinical severity that is correlated with the genotype.


Subject(s)
Exosome Multienzyme Ribonuclease Complex/genetics , Genetic Association Studies , Mutation/genetics , Olivopontocerebellar Atrophies/genetics , RNA-Binding Proteins/genetics , Adolescent , Brain/pathology , Child , Child, Preschool , Cohort Studies , DNA Mutational Analysis , Electromyography , Family Health , Female , Gene Frequency , Genotype , Humans , Infant , Magnetic Resonance Imaging , Male , Olivopontocerebellar Atrophies/complications , Olivopontocerebellar Atrophies/pathology , Young Adult
20.
Science ; 339(6115): 55-9, 2013 Jan 04.
Article in English | MEDLINE | ID: mdl-23288534

ABSTRACT

The future of nanoscale spin-based technologies hinges on a fundamental understanding and dynamic control of atomic-scale magnets. The role of the substrate conduction electrons on the dynamics of supported atomic magnets is still a question of interest lacking experimental insight. We characterized the temperature-dependent dynamical response of artificially constructed magnets, composed of a few exchange-coupled atomic spins adsorbed on a metallic substrate, to spin-polarized currents driven and read out by a magnetic scanning tunneling microscope tip. The dynamics, reflected by two-state spin noise, is quantified by a model that considers the interplay between quantum tunneling and sequential spin transitions driven by electron spin-flip processes and accounts for an observed spin-transfer torque effect.

SELECTION OF CITATIONS
SEARCH DETAIL
...