Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 128
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Am J Hum Genet ; 110(12): 2103-2111, 2023 Dec 07.
Article in English | MEDLINE | ID: mdl-37924809

ABSTRACT

Hereditary spastic parapareses (HSPs) are clinically heterogeneous motor neuron diseases with variable age of onset and severity. Although variants in dozens of genes are implicated in HSPs, much of the genetic basis for pediatric-onset HSP remains unexplained. Here, we re-analyzed clinical exome-sequencing data from siblings with HSP of unknown genetic etiology and identified an inherited nonsense mutation (c.523C>T [p.Arg175Ter]) in the highly conserved RAB1A. The mutation is predicted to produce a truncated protein with an intact RAB GTPase domain but without two C-terminal cysteine residues required for proper subcellular protein localization. Additional RAB1A mutations, including two frameshift mutations and a mosaic missense mutation (c.83T>C [p.Leu28Pro]), were identified in three individuals with similar neurodevelopmental presentations. In rescue experiments, production of the full-length, but not the truncated, RAB1a rescued Golgi structure and cell proliferation in Rab1-depleted cells. In contrast, the missense-variant RAB1a disrupted Golgi structure despite intact Rab1 expression, suggesting a dominant-negative function of the mosaic missense mutation. Knock-down of RAB1A in cultured human embryonic stem cell-derived neurons resulted in impaired neuronal arborization. Finally, RAB1A is located within the 2p14-p15 microdeletion syndrome locus. The similar clinical presentations of individuals with RAB1A loss-of-function mutations and the 2p14-p15 microdeletion syndrome implicate loss of RAB1A in the pathogenesis of neurodevelopmental manifestations of this microdeletion syndrome. Our study identifies a RAB1A-related neurocognitive disorder with speech and motor delay, demonstrates an essential role for RAB1a in neuronal differentiation, and implicates RAB1A in the etiology of the neurodevelopmental sequelae associated with the 2p14-p15 microdeletion syndrome.


Subject(s)
Haploinsufficiency , Spastic Paraplegia, Hereditary , Child , Humans , Haploinsufficiency/genetics , Mutation , Mutation, Missense/genetics , rab GTP-Binding Proteins/genetics , rab GTP-Binding Proteins/metabolism , Golgi Apparatus/metabolism , Spastic Paraplegia, Hereditary/genetics
2.
Cell ; 146(6): 889-903, 2011 Sep 16.
Article in English | MEDLINE | ID: mdl-21925314

ABSTRACT

Complex genomic rearrangements (CGRs) consisting of two or more breakpoint junctions have been observed in genomic disorders. Recently, a chromosome catastrophe phenomenon termed chromothripsis, in which numerous genomic rearrangements are apparently acquired in one single catastrophic event, was described in multiple cancers. Here, we show that constitutionally acquired CGRs share similarities with cancer chromothripsis. In the 17 CGR cases investigated, we observed localization and multiple copy number changes including deletions, duplications, and/or triplications, as well as extensive translocations and inversions. Genomic rearrangements involved varied in size and complexities; in one case, array comparative genomic hybridization revealed 18 copy number changes. Breakpoint sequencing identified characteristic features, including small templated insertions at breakpoints and microhomology at breakpoint junctions, which have been attributed to replicative processes. The resemblance between CGR and chromothripsis suggests similar mechanistic underpinnings. Such chromosome catastrophic events appear to reflect basic DNA metabolism operative throughout an organism's life cycle.


Subject(s)
Chromosome Aberrations , DNA Repair , Developmental Disabilities/genetics , Neoplasms/genetics , Base Sequence , Child , Child, Preschool , Chromosome Breakage , Comparative Genomic Hybridization , DNA Replication , Female , Humans , In Situ Hybridization, Fluorescence , Infant , Male , Molecular Sequence Data
3.
Am J Hum Genet ; 109(12): 2270-2282, 2022 12 01.
Article in English | MEDLINE | ID: mdl-36368327

ABSTRACT

An Xq22.2 region upstream of PLP1 has been proposed to underly a neurological disease trait when deleted in 46,XX females. Deletion mapping revealed that heterozygous deletions encompassing the smallest region of overlap (SRO) spanning six Xq22.2 genes (BEX3, RAB40A, TCEAL4, TCEAL3, TCEAL1, and MORF4L2) associate with an early-onset neurological disease trait (EONDT) consisting of hypotonia, intellectual disability, neurobehavioral abnormalities, and dysmorphic facial features. None of the genes within the SRO have been associated with monogenic disease in OMIM. Through local and international collaborations facilitated by GeneMatcher and Matchmaker Exchange, we have identified and herein report seven de novo variants involving TCEAL1 in seven unrelated families: three hemizygous truncating alleles; one hemizygous missense allele; one heterozygous TCEAL1 full gene deletion; one heterozygous contiguous deletion of TCEAL1, TCEAL3, and TCEAL4; and one heterozygous frameshift variant allele. Variants were identified through exome or genome sequencing with trio analysis or through chromosomal microarray. Comparison with previously reported Xq22 deletions encompassing TCEAL1 identified a more-defined syndrome consisting of hypotonia, abnormal gait, developmental delay/intellectual disability especially affecting expressive language, autistic-like behavior, and mildly dysmorphic facial features. Additional features include strabismus, refractive errors, variable nystagmus, gastroesophageal reflux, constipation, dysmotility, recurrent infections, seizures, and structural brain anomalies. An additional maternally inherited hemizygous missense allele of uncertain significance was identified in a male with hypertonia and spasticity without syndromic features. These data provide evidence that TCEAL1 loss of function causes a neurological rare disease trait involving significant neurological impairment with features overlapping the EONDT phenotype in females with the Xq22 deletion.


Subject(s)
Autistic Disorder , Intellectual Disability , Female , Humans , Male , Autistic Disorder/genetics , Intellectual Disability/genetics , Intellectual Disability/complications , Muscle Hypotonia/genetics , Muscle Hypotonia/complications , Phenotype , Syndrome , Transcription Factors/genetics
4.
J Med Genet ; 60(6): 547-556, 2023 06.
Article in English | MEDLINE | ID: mdl-36150828

ABSTRACT

BACKGROUND: Mosaicism for chromosomal structural abnormalities, other than marker or ring chromosomes, is rarely inherited. METHODS: We performed cytogenetics studies and breakpoint analyses on a family with transmission of mosaicism for a derivative chromosome 8 (der(8)), resulting from an unbalanced translocation between the long arms of chromosomes 8 and 21 over three generations. RESULTS: The proband and his maternal half-sister had mosaicism for a der(8) cell line leading to trisomy of the distal 21q, and both had Down syndrome phenotypic features. Mosaicism for a cell line with the der(8) and a normal cell line was also detected in a maternal half-cousin. The der(8) was inherited from the maternal grandmother who had four abnormal cell lines containing the der(8), in addition to a normal cell line. One maternal half-aunt had the der(8) and an isodicentric chromosome 21 (idic(21)). Sequencing studies revealed microhomologies at the junctures of the der(8) and idic(21) in the half-aunt, suggesting a replicative mechanism in the rearrangement formation. Furthermore, interstitial telomeric sequences (ITS) were identified in the juncture between chromosomes 8 and 21 in the der(8). CONCLUSION: Mosaicism in the proband, his half-sister and half-cousin resulting from loss of chromosome 21 material from the der(8) appears to be a postzygotic event due to the genomic instability of ITS and associated with selective growth advantage of normal cells. The reversion of the inherited der(8) to a normal chromosome 8 in this family resembles revertant mosaicism of point mutations. We propose that ITS could mediate recurring revertant mosaicism for some constitutional chromosomal structural abnormalities.


Subject(s)
Mosaicism , Ring Chromosomes , Humans , Chromosomes, Human, Pair 8/genetics , Karyotyping , In Situ Hybridization, Fluorescence , Chromosome Aberrations , Translocation, Genetic/genetics , Germ Cells
5.
Am J Hum Genet ; 106(1): 129-136, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31883644

ABSTRACT

Birth defects occur in up to 3% of all live births and are the leading cause of infant death. Here we present five individuals from four unrelated families, individuals who share similar phenotypes with disease-causal bi-allelic variants in NADSYN1, encoding NAD synthetase 1, the final enzyme of the nicotinamide adenine dinucleotide (NAD) de novo synthesis pathway. Defects range from the isolated absence of both kidneys to multiple malformations of the vertebrae, heart, limbs, and kidney, and no affected individual survived for more than three months postnatally. NAD is an essential coenzyme for numerous cellular processes. Bi-allelic loss-of-function mutations in genes required for the de novo synthesis of NAD were previously identified in individuals with multiple congenital abnormalities affecting the heart, kidney, vertebrae, and limbs. Functional assessments of NADSYN1 missense variants, through a combination of yeast complementation and enzymatic assays, show impaired enzymatic activity and severely reduced NAD levels. Thus, NADSYN1 represents an additional gene required for NAD synthesis during embryogenesis, and NADSYN1 has bi-allelic missense variants that cause NAD deficiency-dependent malformations. Our findings expand the genotypic spectrum of congenital NAD deficiency disorders and further implicate mutation of additional genes involved in de novo NAD synthesis as potential causes of complex birth defects.


Subject(s)
Carbon-Nitrogen Ligases with Glutamine as Amide-N-Donor/genetics , Congenital Abnormalities/etiology , Multiple Organ Failure/etiology , Mutation, Missense , NAD/deficiency , Alleles , Amino Acid Sequence , Congenital Abnormalities/pathology , Female , Genotype , Gestational Age , Humans , Infant , Infant, Newborn , Male , Multiple Organ Failure/pathology , Pedigree , Phenotype , Pregnancy , Sequence Homology
6.
Am J Med Genet A ; 191(3): 776-785, 2023 03.
Article in English | MEDLINE | ID: mdl-36537114

ABSTRACT

WWOX biallelic loss-of-function pathogenic single nucleotide variants (SNVs) and copy number variants (CNVs) including exonic deletions and duplications cause WWOX-related epileptic encephalopathy (WOREE) syndrome. This disorder is characterized by refractory epilepsy, axial hypotonia, peripheral hypertonia, progressive microcephaly, and premature death. Here we report five patients with WWOX biallelic predicted null variants identified by exome sequencing (ES), genome sequencing (GS), and/or chromosomal microarray analysis (CMA). SNVs and intragenic deletions of one or more exons were commonly reported in WOREE syndrome patients which made the genetic diagnosis challenging and required a combination of different diagnostic technologies. These patients presented with severe, developmental and epileptic encephalopathy (DEE), and other cardinal features consistent with WOREE syndrome. This report expands the clinical phenotype associated with this condition, including failure to thrive in most patients and epilepsy that responded to a ketogenic diet in three patients. Dysmorphic features and abnormal prenatal findings were not commonly observed. Additionally, recurrent pancreatitis and sensorineural hearing loss each were observed in single patients. In summary, these phenotypic features broaden the clinical spectrum of WOREE syndrome.


Subject(s)
Brain Diseases , Epilepsy, Generalized , Epilepsy , Epileptic Syndromes , Female , Pregnancy , Humans , Epilepsy/diagnosis , Epilepsy/genetics , Epileptic Syndromes/genetics , Brain Diseases/genetics , Epilepsy, Generalized/genetics , Exons , WW Domain-Containing Oxidoreductase/genetics , Tumor Suppressor Proteins/genetics
7.
Mol Cell ; 59(6): 956-69, 2015 Sep 17.
Article in English | MEDLINE | ID: mdl-26365382

ABSTRACT

Endosomal protein recycling is a fundamental cellular process important for cellular homeostasis, signaling, and fate determination that is implicated in several diseases. WASH is an actin-nucleating protein essential for this process, and its activity is controlled through K63-linked ubiquitination by the MAGE-L2-TRIM27 ubiquitin ligase. Here, we show that the USP7 deubiquitinating enzyme is an integral component of the MAGE-L2-TRIM27 ligase and is essential for WASH-mediated endosomal actin assembly and protein recycling. Mechanistically, USP7 acts as a molecular rheostat to precisely fine-tune endosomal F-actin levels by counteracting TRIM27 auto-ubiquitination/degradation and preventing overactivation of WASH through directly deubiquitinating it. Importantly, we identify de novo heterozygous loss-of-function mutations of USP7 in individuals with a neurodevelopmental disorder, featuring intellectual disability and autism spectrum disorder. These results provide unanticipated insights into endosomal trafficking, illuminate the cooperativity between an ubiquitin ligase and a deubiquitinating enzyme, and establish a role for USP7 in human neurodevelopmental disease.


Subject(s)
Autism Spectrum Disorder/enzymology , Endosomes/metabolism , Intellectual Disability/enzymology , Microfilament Proteins/metabolism , Ubiquitin Thiolesterase/physiology , Adolescent , Autism Spectrum Disorder/genetics , Child , Child, Preschool , DNA-Binding Proteins/metabolism , Feedback, Physiological , Female , HCT116 Cells , Haploinsufficiency , Humans , Hypothalamus/metabolism , Intellectual Disability/genetics , Male , Neurons/enzymology , Nuclear Proteins/metabolism , Protein Transport , Proteolysis , Sequence Deletion , Ubiquitin-Specific Peptidase 7 , Ubiquitination
8.
Am J Hum Genet ; 105(6): 1262-1273, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31785788

ABSTRACT

It has long been appreciated that genetic analysis of fetal or trophoblast cells in maternal blood could revolutionize prenatal diagnosis. We implemented a protocol for single circulating trophoblast (SCT) testing using positive selection by magnetic-activated cell sorting and single-cell low-coverage whole-genome sequencing to detect fetal aneuploidies and copy-number variants (CNVs) at ∼1 Mb resolution. In 95 validation cases, we identified on average 0.20 putative trophoblasts/mL, of which 55% were of high quality and scorable for both aneuploidy and CNVs. We emphasize the importance of analyzing individual cells because some cells are apoptotic, in S-phase, or otherwise of poor quality. When two or more high-quality trophoblast cells were available for singleton pregnancies, there was complete concordance between all trophoblasts unless there was evidence of confined placental mosaicism. SCT results were highly concordant with available clinical data from chorionic villus sampling (CVS) or amniocentesis procedures. Although determining the exact sensitivity and specificity will require more data, this study further supports the potential for SCT testing to become a diagnostic prenatal test.


Subject(s)
Chromosome Disorders/diagnosis , Genetic Markers , Noninvasive Prenatal Testing/methods , Placenta/metabolism , Trophoblasts/cytology , Trophoblasts/metabolism , Adult , Chromosome Disorders/genetics , DNA Copy Number Variations , Female , Humans , Male , Placenta/cytology , Pregnancy , Single-Cell Analysis , Young Adult
9.
Am J Hum Genet ; 105(6): 1237-1253, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31785787

ABSTRACT

We report an early-onset autosomal-recessive neurological disease with cerebellar atrophy and lysosomal dysfunction. We identified bi-allelic loss-of-function (LoF) variants in Oxidative Resistance 1 (OXR1) in five individuals from three families; these individuals presented with a history of severe global developmental delay, current intellectual disability, language delay, cerebellar atrophy, and seizures. While OXR1 is known to play a role in oxidative stress resistance, its molecular functions are not well established. OXR1 contains three conserved domains: LysM, GRAM, and TLDc. The gene encodes at least six transcripts, including some that only consist of the C-terminal TLDc domain. We utilized Drosophila to assess the phenotypes associated with loss of mustard (mtd), the fly homolog of OXR1. Strong LoF mutants exhibit late pupal lethality or pupal eclosion defects. Interestingly, although mtd encodes 26 transcripts, severe LoF and null mutations can be rescued by a single short human OXR1 cDNA that only contains the TLDc domain. Similar rescue is observed with the TLDc domain of NCOA7, another human homolog of mtd. Loss of mtd in neurons leads to massive cell loss, early death, and an accumulation of aberrant lysosomal structures, similar to what we observe in fibroblasts of affected individuals. Our data indicate that mtd and OXR1 are required for proper lysosomal function; this is consistent with observations that NCOA7 is required for lysosomal acidification.


Subject(s)
Atrophy/pathology , Cerebellar Diseases/pathology , Lysosomes/pathology , Mitochondrial Proteins/metabolism , Nervous System Diseases/pathology , Oxidative Stress , Adolescent , Adult , Animals , Atrophy/genetics , Atrophy/metabolism , Cerebellar Diseases/genetics , Cerebellar Diseases/metabolism , Child , Drosophila melanogaster/growth & development , Drosophila melanogaster/metabolism , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Humans , Lysosomes/metabolism , Male , Mitochondrial Proteins/genetics , Nervous System Diseases/genetics , Nervous System Diseases/metabolism , Pedigree , Phenotype , Young Adult
10.
Am J Hum Genet ; 104(3): 422-438, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30773277

ABSTRACT

SPONASTRIME dysplasia is an autosomal-recessive spondyloepimetaphyseal dysplasia characterized by spine (spondylar) abnormalities, midface hypoplasia with a depressed nasal bridge, metaphyseal striations, and disproportionate short stature. Scoliosis, coxa vara, childhood cataracts, short dental roots, and hypogammaglobulinemia have also been reported in this disorder. Although an autosomal-recessive inheritance pattern has been hypothesized, pathogenic variants in a specific gene have not been discovered in individuals with SPONASTRIME dysplasia. Here, we identified bi-allelic variants in TONSL, which encodes the Tonsoku-like DNA repair protein, in nine subjects (from eight families) with SPONASTRIME dysplasia, and four subjects (from three families) with short stature of varied severity and spondylometaphyseal dysplasia with or without immunologic and hematologic abnormalities, but no definitive metaphyseal striations at diagnosis. The finding of early embryonic lethality in a Tonsl-/- murine model and the discovery of reduced length, spinal abnormalities, reduced numbers of neutrophils, and early lethality in a tonsl-/- zebrafish model both support the hypomorphic nature of the identified TONSL variants. Moreover, functional studies revealed increased amounts of spontaneous replication fork stalling and chromosomal aberrations, as well as fewer camptothecin (CPT)-induced RAD51 foci in subject-derived cell lines. Importantly, these cellular defects were rescued upon re-expression of wild-type (WT) TONSL; this rescue is consistent with the hypothesis that hypomorphic TONSL variants are pathogenic. Overall, our studies in humans, mice, zebrafish, and subject-derived cell lines confirm that pathogenic variants in TONSL impair DNA replication and homologous recombination-dependent repair processes, and they lead to a spectrum of skeletal dysplasia phenotypes with numerous extra-skeletal manifestations.


Subject(s)
Chromosomal Instability , DNA Damage , Genetic Variation , Musculoskeletal Abnormalities/pathology , NF-kappa B/genetics , Osteochondrodysplasias/pathology , Adolescent , Adult , Alleles , Animals , Cells, Cultured , Child , Child, Preschool , Female , Fibroblasts/metabolism , Fibroblasts/pathology , Genetic Association Studies , Humans , Mice , Mice, Knockout , Musculoskeletal Abnormalities/genetics , Osteochondrodysplasias/genetics , Exome Sequencing , Young Adult , Zebrafish
11.
Genet Med ; 24(2): 364-373, 2022 02.
Article in English | MEDLINE | ID: mdl-34906496

ABSTRACT

PURPOSE: BRG1/BRM-associated factor (BAF) complex is a chromatin remodeling complex that plays a critical role in gene regulation. Defects in the genes encoding BAF subunits lead to BAFopathies, a group of neurodevelopmental disorders with extensive locus and phenotypic heterogeneity. METHODS: We retrospectively analyzed data from 16,243 patients referred for clinical exome sequencing (ES) with a focus on the BAF complex. We applied a genotype-first approach, combining predicted genic constraints to propose candidate BAFopathy genes. RESULTS: We identified 127 patients carrying pathogenic variants, likely pathogenic variants, or de novo variants of unknown clinical significance in 11 known BAFopathy genes. Those include 34 patients molecularly diagnosed using ES reanalysis with new gene-disease evidence (n = 21) or variant reclassifications in known BAFopathy genes (n = 13). We also identified de novo or predicted loss-of-function variants in 4 candidate BAFopathy genes, including ACTL6A, BICRA (implicated in Coffin-Siris syndrome during this study), PBRM1, and SMARCC1. CONCLUSION: We report the mutational spectrum of BAFopathies in an ES cohort. A genotype-driven and pathway-based reanalysis of ES data identified new evidence for candidate genes involved in BAFopathies. Further mechanistic and phenotypic characterization of additional patients are warranted to confirm their roles in human disease and to delineate their associated phenotypic spectrums.


Subject(s)
Abnormalities, Multiple , Hand Deformities, Congenital , Micrognathism , Abnormalities, Multiple/genetics , Actins/genetics , Chromosomal Proteins, Non-Histone/genetics , DNA-Binding Proteins/genetics , Exome/genetics , Hand Deformities, Congenital/genetics , Humans , Micrognathism/genetics , Retrospective Studies
12.
Mol Psychiatry ; 26(5): 1706-1718, 2021 05.
Article in English | MEDLINE | ID: mdl-33597717

ABSTRACT

Mendelian and early-onset severe psychiatric phenotypes often involve genetic variants having a large effect, offering opportunities for genetic discoveries and early therapeutic interventions. Here, the index case is an 18-year-old boy, who at 14 years of age had a decline in cognitive functioning over the course of a year and subsequently presented with catatonia, auditory and visual hallucinations, paranoia, aggression, mood dysregulation, and disorganized thoughts. Exome sequencing revealed a stop-gain mutation in RCL1 (NM_005772.4:c.370 C > T, p.Gln124Ter), encoding an RNA 3'-terminal phosphate cyclase-like protein that is highly conserved across eukaryotic species. Subsequent investigations across two academic medical centers identified eleven additional cases of RCL1 copy number variations (CNVs) with varying neurodevelopmental or psychiatric phenotypes. These findings suggest that dosage variation of RCL1 contributes to a range of neurological and clinical phenotypes.


Subject(s)
DNA Copy Number Variations , Adolescent , DNA Copy Number Variations/genetics , Humans , Male , Mutation/genetics , Phenotype , Exome Sequencing
13.
Am J Med Genet A ; 188(11): 3184-3190, 2022 11.
Article in English | MEDLINE | ID: mdl-36065636

ABSTRACT

Stroke causes significant disability and is a common cause of death worldwide. Previous studies have estimated that 1%-5% of stroke is attributable to monogenic etiologies. We set out to assess the utility of clinical exome sequencing (ES) in the evaluation of stroke. We retrospectively analyzed 124 individuals who received ES at the Baylor Genetics reference lab between 2012 and 2021 who had stroke as a major part of their reported phenotype. Ages ranged from 10 days to 69 years. 8.9% of the cohort received a diagnosis, including 25% of infants less than 1 year old; an additional 10.5% of the cohort received a probable diagnosis. We identified several syndromes that predispose to stroke such as COL4A1-related brain small vessel disease, homocystinuria caused by CBS mutation, POLG-related disorders, TTC19-linked mitochondrial disease, and RNASEH2A associated Aicardi-Goutieres syndrome. We also observed pathogenic variants in NSD1, PKHD1, HRAS, and ATP13A2, which are genes rarely associated with stroke. Although stroke is a complex phenotype with varying pathologies and risk factors, these results show that use of exome sequencing can be highly relevant in stroke, especially for those presenting <1 year of age.


Subject(s)
Exome , Stroke , Exome/genetics , Humans , Phenotype , Retrospective Studies , Stroke/diagnosis , Stroke/genetics , Exome Sequencing/methods
14.
Am J Med Genet A ; 188(6): 1858-1862, 2022 06.
Article in English | MEDLINE | ID: mdl-35188328

ABSTRACT

Leiomodin-2 (LMOD2) is an important regulator of the thin filament length, known to promote elongation of actin through polymerization at pointed ends. Mice with Lmod2 deficiency die around 3 weeks of age due to severe dilated cardiomyopathy (DCM), resulting from decreased heart contractility due to shorter thin filaments. To date, there have been three infants from two families reported with biallelic variants in LMOD2, presenting with perinatal onset DCM. Here, we describe a third family with a child harboring a previously described homozygous frameshift variant, c.1243_1244delCT (p.L415Vfs*108) with DCM, presenting later in infancy at 9 months of age. Family history was relevant for a sibling who died suddenly at 1 year of age after being diagnosed with cardiomegaly. LMOD2-related cardiomyopathy is a rare form of inherited cardiomyopathy resulting from thin filament length dysregulation and should be considered in genetic evaluation of newborns and infants with suspected autosomal recessive inheritance or sporadic early onset cardiomyopathy.


Subject(s)
Cardiomyopathies , Cardiomyopathy, Dilated , Actin Cytoskeleton/genetics , Animals , Cardiomyopathy, Dilated/diagnosis , Cardiomyopathy, Dilated/genetics , Cytoskeletal Proteins/genetics , Heart , Humans , Infant, Newborn , Mice , Muscle Proteins/genetics , Sarcomeres
15.
Am J Med Genet A ; 188(12): 3492-3504, 2022 12.
Article in English | MEDLINE | ID: mdl-36135330

ABSTRACT

Esophageal atresia/tracheoesophageal fistula (EA/TEF) is a life-threatening birth defect that often occurs with other major birth defects (EA/TEF+). Despite advances in genetic testing, a molecular diagnosis can only be made in a minority of EA/TEF+ cases. Here, we analyzed clinical exome sequencing data and data from the DECIPHER database to determine the efficacy of exome sequencing in cases of EA/TEF+ and to identify phenotypic expansions involving EA/TEF. Among 67 individuals with EA/TEF+ referred for clinical exome sequencing, a definitive or probable diagnosis was made in 11 cases for an efficacy rate of 16% (11/67). This efficacy rate is significantly lower than that reported for other major birth defects, suggesting that polygenic, multifactorial, epigenetic, and/or environmental factors may play a particularly important role in EA/TEF pathogenesis. Our cohort included individuals with pathogenic or likely pathogenic variants that affect TCF4 and its downstream target NRXN1, and FANCA, FANCB, and FANCC, which are associated with Fanconi anemia. These cases, previously published case reports, and comparisons to other EA/TEF genes made using a machine learning algorithm, provide evidence in support of a potential pathogenic role for these genes in the development of EA/TEF.


Subject(s)
Esophageal Atresia , Tracheoesophageal Fistula , Humans , Tracheoesophageal Fistula/diagnosis , Tracheoesophageal Fistula/genetics , Tracheoesophageal Fistula/complications , Esophageal Atresia/diagnosis , Esophageal Atresia/genetics , Esophageal Atresia/complications , Exome/genetics , Exome Sequencing
16.
Genet Med ; 23(7): 1234-1245, 2021 07.
Article in English | MEDLINE | ID: mdl-33824499

ABSTRACT

PURPOSE: Proline Rich 12 (PRR12) is a gene of unknown function with suspected DNA-binding activity, expressed in developing mice and human brains. Predicted loss-of-function variants in this gene are extremely rare, indicating high intolerance of haploinsufficiency. METHODS: Three individuals with intellectual disability and iris anomalies and truncating de novo PRR12 variants were described previously. We add 21 individuals with similar PRR12 variants identified via matchmaking platforms, bringing the total number to 24. RESULTS: We observed 12 frameshift, 6 nonsense, 1 splice-site, and 2 missense variants and one patient with a gross deletion involving PRR12. Three individuals had additional genetic findings, possibly confounding the phenotype. All patients had developmental impairment. Variable structural eye defects were observed in 12/24 individuals (50%) including anophthalmia, microphthalmia, colobomas, optic nerve and iris abnormalities. Additional common features included hypotonia (61%), heart defects (52%), growth failure (54%), and kidney anomalies (35%). PrediXcan analysis showed that phecodes most strongly associated with reduced predicted PRR12 expression were enriched for eye- (7/30) and kidney- (4/30) phenotypes, such as wet macular degeneration and chronic kidney disease. CONCLUSION: These findings support PRR12 haploinsufficiency as a cause for a novel disorder with a wide clinical spectrum marked chiefly by neurodevelopmental and eye abnormalities.


Subject(s)
Haploinsufficiency , Intellectual Disability , Animals , Haploinsufficiency/genetics , Humans , Intellectual Disability/genetics , Mice , Muscle Hypotonia , Mutation, Missense , Phenotype
17.
Clin Genet ; 100(2): 227-233, 2021 08.
Article in English | MEDLINE | ID: mdl-33963760

ABSTRACT

PPP3CA encodes the catalytic subunit of calcineurin, a calcium-calmodulin-regulated serine-threonine phosphatase. Loss-of-function (LoF) variants in the catalytic domain have been associated with epilepsy, while gain-of-function (GoF) variants in the auto-inhibitory domain cause multiple congenital abnormalities. We herein report five new patients with de novo PPP3CA variants. Interestingly, the two frameshift variants in this study and the six truncating variants reported previously are all located within a 26-amino acid region in the regulatory domain (RD). Patients with a truncating variant had more severe earlier onset seizures compared to patients with a LoF missense variant, while autism spectrum disorder was a more frequent feature in the latter. Expression studies of a truncating variant showed apparent RNA expression from the mutant allele, but no detectable mutant protein. Our data suggest that PPP3CA truncating variants clustered in the RD, causing more severe early-onset refractory epilepsy and representing a type of variants distinct from LoF or GoF missense variants.


Subject(s)
Calcineurin/genetics , Epilepsy/genetics , Mutation , Adolescent , Calcineurin/metabolism , Child , Child, Preschool , Drug Resistant Epilepsy/etiology , Drug Resistant Epilepsy/genetics , Epilepsy/etiology , Female , Gene Expression , Humans , Male , Sequence Analysis, RNA
18.
Am J Med Genet A ; 185(10): 2903-2912, 2021 10.
Article in English | MEDLINE | ID: mdl-34061437

ABSTRACT

Trisomy 16 is the most common autosomal trisomy in humans, which is almost uniformly embryonic lethal. Partial trisomy 16 including a segment of the long arm of chromosome 16 is occasionally compatible with life and has been associated with severe congenital defects, growth retardation, and early lethality. Segmental trisomy of 16q is usually described concomitantly with partial monosomy of another chromosome, often resulting from a parental balanced translocation. Pure partial chromosome 16q trisomy is exceedingly rare. About nine children with 16q12→qter and 16q13→qter duplication have been reported in the literature, almost all described with monosomy of a second chromosome, and highlighting very few long-term survivors. A single individual with pure partial distal 16q12.1q23.3 duplication has been reported in an infant, underscoring complexities of genetic counseling and management, especially in view of life-limiting congenital anomalies in rare survivors. Here, we present a 12-month-old child with pure 16q12.2q24.3 trisomy, having continued morbidity related to pulmonary hypertension and chronic lung disease. The features of intrauterine growth retardation, facial dysmorphism, hypotonia, congenital heart defect, distal contractures, urogenital abnormalities, and hearing loss support the association with 16q partial trisomy, as in previous studies. This report expands our current understanding related to the survival of infants with large segmental aneusomy of the long arm of chromosome 16.


Subject(s)
Congenital Abnormalities/genetics , Heart Defects, Congenital/genetics , Translocation, Genetic , Trisomy/genetics , Child , Chromosomes, Human, Pair 16/genetics , Congenital Abnormalities/pathology , Heart Defects, Congenital/pathology , Humans , Hypertension, Pulmonary/complications , Hypertension, Pulmonary/genetics , Hypertension, Pulmonary/pathology , Infant , Karyotyping , Lung Diseases/complications , Lung Diseases/genetics , Lung Diseases/pathology , Male , Mosaicism , Trisomy/pathology
19.
Am J Med Genet A ; 185(5): 1388-1398, 2021 05.
Article in English | MEDLINE | ID: mdl-33576134

ABSTRACT

Distal 1q21.1 microdeletions have shown highly variable clinical expressivity and incomplete penetrance, with affected individuals manifesting a broad spectrum of nonspecific features. The goals of this study were to better describe the phenotypic spectrum of patients with distal 1q21.1 microdeletions and to compare the clinical features among affected individuals. We performed a retrospective chart review of 47 individuals with distal 1q21.1 microdeletions tested at a large clinical genetic testing laboratory, with most patients being clinically evaluated in the same children's hospital. Health information such as growth charts, results of imaging studies, developmental history, and progress notes were collected. Statistical analysis was performed using Fisher's exact test to compare clinical features among study subjects. Common features in our cohort include microcephaly (51.2%), seizures (29.8%), developmental delay (74.5%), failure to thrive (FTT) (68.1%), dysmorphic features (63.8%), and a variety of congenital anomalies such as cardiac abnormalities (23.4%) and genitourinary abnormalities (19.1%). Compared to prior literature, we found that seizures, brain anomalies, and FTT were more prevalent among our study cohort. Females were more likely than males to have microcephaly (p = 0.0199) and cardiac abnormalities (p = 0.0018). Based on existing genome-wide clinical testing results, at least a quarter of the cohort had additional genetic findings that may impact the phenotype of the individual. Our study represents the largest cohort of distal 1q21.1 microdeletion carriers available in the literature thus far, and it further illustrates the wide spectrum of clinical manifestations among symptomatic individuals. These results may allow for improved genetic counseling and management of affected individuals. Future studies may help to elucidate the underlying molecular mechanisms impacting the phenotypic variability observed with this microdeletion.


Subject(s)
Abnormalities, Multiple/genetics , Heart Defects, Congenital/genetics , Intellectual Disability/genetics , Megalencephaly/genetics , Microcephaly/genetics , Abnormalities, Multiple/diagnosis , Abnormalities, Multiple/physiopathology , Adolescent , Adult , Child , Child, Preschool , Chromosome Deletion , Chromosomes, Human, Pair 1/genetics , DNA Copy Number Variations/genetics , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/physiopathology , Failure to Thrive/complications , Failure to Thrive/genetics , Failure to Thrive/physiopathology , Female , Genetic Counseling , Genetic Testing/methods , Heart Defects, Congenital/complications , Heart Defects, Congenital/diagnosis , Heart Defects, Congenital/physiopathology , Humans , Infant , Intellectual Disability/complications , Intellectual Disability/diagnosis , Intellectual Disability/physiopathology , Male , Megalencephaly/complications , Megalencephaly/diagnosis , Megalencephaly/physiopathology , Microcephaly/complications , Microcephaly/diagnosis , Microcephaly/physiopathology , Pedigree , Seizures/complications , Seizures/genetics , Seizures/physiopathology , Young Adult
20.
Epilepsia ; 62(7): e103-e109, 2021 07.
Article in English | MEDLINE | ID: mdl-34041744

ABSTRACT

CSNK2B has recently been implicated as a disease gene for neurodevelopmental disability (NDD) and epilepsy. Information about developmental outcomes has been limited by the young age and short follow-up for many of the previously reported cases, and further delineation of the spectrum of associated phenotypes is needed. We present 25 new patients with variants in CSNK2B and refine the associated NDD and epilepsy phenotypes. CSNK2B variants were identified by research or clinical exome sequencing, and investigators from different centers were connected via GeneMatcher. Most individuals had developmental delay and generalized epilepsy with onset in the first 2 years. However, we found a broad spectrum of phenotypic severity, ranging from early normal development with pharmacoresponsive seizures to profound intellectual disability with intractable epilepsy and recurrent refractory status epilepticus. These findings suggest that CSNK2B should be considered in the diagnostic evaluation of patients with a broad range of NDD with treatable or intractable seizures.


Subject(s)
Developmental Disabilities/genetics , Epilepsy, Generalized/genetics , Adolescent , Adult , Age of Onset , Child , Child, Preschool , Developmental Disabilities/physiopathology , Epilepsies, Myoclonic/diagnosis , Epilepsies, Myoclonic/etiology , Epilepsies, Myoclonic/genetics , Epilepsy, Generalized/diagnosis , Epilepsy, Generalized/etiology , Exome/genetics , Female , Genetic Variation , Humans , Infant , Intellectual Disability/etiology , Intellectual Disability/genetics , Male , Mutation/genetics , Phenotype , Status Epilepticus/diagnosis , Status Epilepticus/etiology , Status Epilepticus/genetics , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL