Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 65
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Nat Rev Genet ; 25(6): 401-415, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38238519

ABSTRACT

Genomic technologies, such as targeted, exome and short-read genome sequencing approaches, have revolutionized the care of patients with rare genetic diseases. However, more than half of patients remain without a diagnosis. Emerging approaches from research-based settings such as long-read genome sequencing and optical genome mapping hold promise for improving the identification of disease-causal genetic variants. In addition, new omic technologies that measure the transcriptome, epigenome, proteome or metabolome are showing great potential for variant interpretation. As genetic testing options rapidly expand, the clinical community needs to be mindful of their individual strengths and limitations, as well as remaining challenges, to select the appropriate diagnostic test, correctly interpret results and drive innovation to address insufficiencies. If used effectively - through truly integrative multi-omics approaches and data sharing - the resulting large quantities of data from these established and emerging technologies will greatly improve the interpretative power of genetic and genomic diagnostics for rare diseases.


Subject(s)
Genetic Testing , Genomics , Rare Diseases , Humans , Rare Diseases/genetics , Rare Diseases/diagnosis , Genetic Testing/methods , Genomics/methods , Genetic Diseases, Inborn/diagnosis , Genetic Diseases, Inborn/genetics
2.
Am J Hum Genet ; 109(11): 1947-1959, 2022 11 03.
Article in English | MEDLINE | ID: mdl-36332610

ABSTRACT

The past decade has witnessed a rapid evolution in rare disease (RD) research, fueled by the availability of genome-wide (exome and genome) sequencing. In 2011, as this transformative technology was introduced to the research community, the Care4Rare Canada Consortium was launched: initially as FORGE, followed by Care4Rare, and Care4Rare SOLVE. Over what amounted to three eras of diagnosis and discovery, the Care4Rare Consortium used exome sequencing and, more recently, genome and other 'omic technologies to identify the molecular cause of unsolved RDs. We achieved a diagnostic yield of 34% (623/1,806 of participating families), including the discovery of deleterious variants in 121 genes not previously associated with disease, and we continue to study candidate variants in novel genes for 145 families. The Consortium has made significant contributions to RD research, including development of platforms for data collection and sharing and instigating a Canadian network to catalyze functional characterization research of novel genes. The Consortium was instrumental to implementing genome-wide sequencing as a publicly funded test for RD diagnosis in Canada. Despite the successes of the past decade, the challenge of solving all RDs remains enormous, and the work is far from over. We must leverage clinical and 'omic data for secondary use, develop tools and policies to support safe data sharing, continue to explore the utility of new and emerging technologies, and optimize research protocols to delineate complex disease mechanisms. Successful approaches in each of these realms is required to offer diagnostic clarity to all families with RDs.


Subject(s)
Exome , Rare Diseases , Humans , Rare Diseases/diagnosis , Rare Diseases/genetics , Canada , Exome/genetics , Exome Sequencing , Genetic Association Studies
3.
Am J Hum Genet ; 109(10): 1923-1931, 2022 10 06.
Article in English | MEDLINE | ID: mdl-36067766

ABSTRACT

MTSS2, also known as MTSS1L, binds to plasma membranes and modulates their bending. MTSS2 is highly expressed in the central nervous system (CNS) and appears to be involved in activity-dependent synaptic plasticity. Variants in MTSS2 have not yet been associated with a human phenotype in OMIM. Here we report five individuals with the same heterozygous de novo variant in MTSS2 (GenBank: NM_138383.2: c.2011C>T [p.Arg671Trp]) identified by exome sequencing. The individuals present with global developmental delay, mild intellectual disability, ophthalmological anomalies, microcephaly or relative microcephaly, and shared mild facial dysmorphisms. Immunoblots of fibroblasts from two affected individuals revealed that the variant does not significantly alter MTSS2 levels. We modeled the variant in Drosophila and showed that the fly ortholog missing-in-metastasis (mim) was widely expressed in most neurons and a subset of glia of the CNS. Loss of mim led to a reduction in lifespan, impaired locomotor behavior, and reduced synaptic transmission in adult flies. Expression of the human MTSS2 reference cDNA rescued the mim loss-of-function (LoF) phenotypes, whereas the c.2011C>T variant had decreased rescue ability compared to the reference, suggesting it is a partial LoF allele. However, elevated expression of the variant, but not the reference MTSS2 cDNA, led to similar defects as observed by mim LoF, suggesting that the variant is toxic and may act as a dominant-negative allele when expressed in flies. In summary, our findings support that mim is important for appropriate neural function, and that the MTSS2 c.2011C>T variant causes a syndromic form of intellectual disability.


Subject(s)
Intellectual Disability , Microcephaly , Nervous System Malformations , Animals , DNA, Complementary , Drosophila/genetics , Humans , Intellectual Disability/genetics , Intellectual Disability/pathology , Membrane Proteins , Microcephaly/genetics , Microfilament Proteins , Mutation, Missense/genetics , Nervous System Malformations/genetics , Phenotype
4.
Hum Mol Genet ; 31(4): 614-624, 2022 02 21.
Article in English | MEDLINE | ID: mdl-34542157

ABSTRACT

SHQ1 is essential for biogenesis of H/ACA ribonucleoproteins, a class of molecules important for processing ribosomal RNAs, modifying spliceosomal small nuclear RNAs and stabilizing telomerase. Components of the H/ACA ribonucleoprotein complex have been linked to neurological developmental defects. Here, we report two sibling pairs from unrelated families with compound heterozygous variants in SHQ1. Exome sequencing was used to detect disease causing variants, which were submitted to 'matching' platforms linked to MatchMaker Exchange. Phenotype comparisons supported these matches. The affected individuals present with early-onset dystonia, with individuals from one family displaying additional neurological phenotypes, including neurodegeneration. As a result of cerebrospinal fluid studies suggesting possible abnormal dopamine metabolism, a trial of levodopa replacement therapy was started but no clear response was noted. We show that fibroblasts from affected individuals have dramatic loss of SHQ1 protein. Variants from both families were expressed in Saccharomyces cerevisiae, resulting in a strong reduction in H/ACA snoRNA production and remarkable defects in rRNA processing and ribosome formation. Our study identifies SHQ1 as associated with neurological disease, including early-onset dystonia, and begins to delineate the molecular etiology of this novel condition.


Subject(s)
Dystonia , Dystonic Disorders , Intracellular Signaling Peptides and Proteins , Saccharomyces cerevisiae Proteins , Dystonia/genetics , Dystonic Disorders/genetics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/metabolism , Nuclear Proteins/genetics , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae/metabolism , Saccharomyces cerevisiae Proteins/genetics
5.
Am J Hum Genet ; 108(10): 2017-2023, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34587489

ABSTRACT

ABHD16A (abhydrolase domain-containing protein 16A, phospholipase) encodes the major phosphatidylserine (PS) lipase in the brain. PS lipase synthesizes lysophosphatidylserine, an important signaling lipid that functions in the mammalian central nervous system. ABHD16A has not yet been associated with a human disease. In this report, we present a cohort of 11 affected individuals from six unrelated families with a complicated form of hereditary spastic paraplegia (HSP) who carry bi-allelic deleterious variants in ABHD16A. Affected individuals present with a similar phenotype consisting of global developmental delay/intellectual disability, progressive spasticity affecting the upper and lower limbs, and corpus callosum and white matter anomalies. Immunoblot analysis on extracts from fibroblasts from four affected individuals demonstrated little to no ABHD16A protein levels compared to controls. Our findings add ABHD16A to the growing list of lipid genes in which dysregulation can cause complicated forms of HSP and begin to describe the molecular etiology of this condition.


Subject(s)
Cerebral Palsy/pathology , Intellectual Disability/pathology , Leukoencephalopathies/pathology , Monoacylglycerol Lipases/genetics , Mutation , Spastic Paraplegia, Hereditary/pathology , Adolescent , Adult , Cerebral Palsy/etiology , Cerebral Palsy/metabolism , Child , Child, Preschool , Cohort Studies , Female , Humans , Intellectual Disability/etiology , Intellectual Disability/metabolism , Leukoencephalopathies/etiology , Leukoencephalopathies/metabolism , Male , Monoacylglycerol Lipases/deficiency , Pedigree , Phenotype , Spastic Paraplegia, Hereditary/etiology , Spastic Paraplegia, Hereditary/metabolism , Young Adult
6.
Am J Hum Genet ; 108(4): 749-756, 2021 04 01.
Article in English | MEDLINE | ID: mdl-33743206

ABSTRACT

The DNA damage-binding protein 1 (DDB1) is part of the CUL4-DDB1 ubiquitin E3 ligase complex (CRL4), which is essential for DNA repair, chromatin remodeling, DNA replication, and signal transduction. Loss-of-function variants in genes encoding the complex components CUL4 and PHIP have been reported to cause syndromic intellectual disability with hypotonia and obesity, but no phenotype has been reported in association with DDB1 variants. Here, we report eight unrelated individuals, identified through Matchmaker Exchange, with de novo monoallelic variants in DDB1, including one recurrent variant in four individuals. The affected individuals have a consistent phenotype of hypotonia, mild to moderate intellectual disability, and similar facies, including horizontal or slightly bowed eyebrows, deep-set eyes, full cheeks, a short nose, and large, fleshy and forward-facing earlobes, demonstrated in the composite face generated from the cohort. Digital anomalies, including brachydactyly and syndactyly, were common. Three older individuals have obesity. We show that cells derived from affected individuals have altered DDB1 function resulting in abnormal DNA damage signatures and histone methylation following UV-induced DNA damage. Overall, our study adds to the growing family of neurodevelopmental phenotypes mediated by disruption of the CRL4 ubiquitin ligase pathway and begins to delineate the phenotypic and molecular effects of DDB1 misregulation.


Subject(s)
Alleles , DNA Repair/genetics , DNA-Binding Proteins/genetics , Mutation , Neurodevelopmental Disorders/genetics , Adolescent , Child , Child, Preschool , Female , Humans , Male , Phenotype , Syndrome
7.
J Hum Genet ; 69(2): 101-105, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37904029

ABSTRACT

Partial duplications of genes can be challenging to detect and interpret and, therefore, likely represent an underreported cause of human disease. X-linked dominant variants in ATRX are associated with Alpha-thalassemia/impaired intellectual development syndrome, X-linked (ATR-X syndrome), a clinically heterogeneous disease generally presenting with intellectual disability, hypotonia, characteristic facies, genital anomalies, and alpha-thalassemia. We describe an affected male with a de novo hemizygous intragenic duplication of ~43.6 kb in ATRX, detected by research genome sequencing following non-diagnostic clinical testing. RNA sequencing and DNA methylation episignature analyses were central in variant interpretation, and this duplication was subsequently interpreted as disease-causing. This represents the smallest reported tandem duplication within ATRX associated with disease. This case demonstrates the diagnostic utility of integrating multiple omics technologies, which can ultimately lead to a definitive diagnosis for rare disease patients.


Subject(s)
Intellectual Disability , Mental Retardation, X-Linked , alpha-Thalassemia , Humans , Male , alpha-Thalassemia/diagnosis , alpha-Thalassemia/genetics , X-linked Nuclear Protein/genetics , DNA Copy Number Variations/genetics , Mental Retardation, X-Linked/diagnosis , Mental Retardation, X-Linked/genetics , Intellectual Disability/diagnosis , Intellectual Disability/genetics
8.
Am J Med Genet A ; 194(3): e63455, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37921537

ABSTRACT

Our understanding of genetic and phenotypic heterogeneity associated with the clinical spectrum of rare diseases continues to expand. Thorough phenotypic descriptions and model organism functional studies are valuable tools in dissecting the biology of the disease process. Kinesin genes are well known to be associated with specific disease phenotypes and a subset of kinesin genes, including KIF21A, have been associated with more than one disease. Here we report two patients with KIF21A variants identified by exome sequencing; one with biallelic variants, supporting a novel KIF21A related syndrome with recessive inheritance and the second report of this condition, and another with a heterozygous de novo variant allele representing a phenotypic expansion of the condition described to date. We provide detailed phenotypic information on both families, including a novel neuropathology finding of neuroaxonal dystrophy associated with biallelic variants in KIF21A. Additionally, we studied the dominant variant in Saccharomyces cerevisiae to assess variant pathogenicity and found that this variant appears to impair protein function. KIF21A associated disease has mounting evidence for phenotypic heterogeneity; further patients and study of an allelic series are required to define the phenotypic spectrum and further explore the molecular etiology for each of these conditions.


Subject(s)
Kinesins , Nervous System Diseases , Humans , Kinesins/genetics , Phenotype , Mutation
9.
Am J Med Genet A ; 194(3): e63466, 2024 Mar.
Article in English | MEDLINE | ID: mdl-37949664

ABSTRACT

Activating variants in the PIK3CA gene cause a heterogeneous spectrum of disorders that involve congenital or early-onset segmental/focal overgrowth, now referred to as PIK3CA-related overgrowth spectrum (PROS). Historically, the clinical diagnoses of patients with PROS included a range of distinct syndromes, including CLOVES syndrome, dysplastic megalencephaly, hemimegalencephaly, focal cortical dysplasia, Klippel-Trenaunay syndrome, CLAPO syndrome, fibroadipose hyperplasia or overgrowth, hemihyperplasia multiple lipomatosis, and megalencephaly capillary malformation-polymicrogyria (MCAP) syndrome. MCAP is a sporadic overgrowth disorder that exhibits core features of progressive megalencephaly, vascular malformations, distal limb malformations, cortical brain malformations, and connective tissue dysplasia. In 2012, our research group contributed to the identification of predominantly mosaic, gain-of-function variants in PIK3CA as an underlying genetic cause of the syndrome. Mosaic variants are technically more difficult to detect and require implementation of more sensitive sequencing technologies and less stringent variant calling algorithms. In this study, we demonstrated the utility of deep sequencing using the Illumina TruSight Oncology 500 (TSO500) sequencing panel in identifying variants with low allele fractions in a series of patients with PROS and suspected mosaicism: pathogenic, mosaic PIK3CA variants were identified in all 13 individuals, including 6 positive controls. This study highlights the importance of screening for low-level mosaic variants in PROS patients. The use of targeted panels with deep sequencing in clinical genetic testing laboratories would improve diagnostic yield and accuracy within this patient population.


Subject(s)
Abnormalities, Multiple , Megalencephaly , Musculoskeletal Abnormalities , Skin Diseases, Vascular , Telangiectasis/congenital , Vascular Malformations , Humans , Mutation , Musculoskeletal Abnormalities/genetics , Class I Phosphatidylinositol 3-Kinases/genetics , Vascular Malformations/diagnosis , Vascular Malformations/genetics , High-Throughput Nucleotide Sequencing
10.
Am J Med Genet A ; 194(5): e63522, 2024 May.
Article in English | MEDLINE | ID: mdl-38131126

ABSTRACT

Despite significant advancements in rare genetic disease diagnostics, many patients with rare genetic disease remain without a molecular diagnosis. Novel tools and methods are needed to improve the detection of disease-associated variants and understand the genetic basis of many rare diseases. Long-read genome sequencing provides improved sequencing in highly repetitive, homologous, and low-complexity regions, and improved assessment of structural variation and complex genomic rearrangements compared to short-read genome sequencing. As such, it is a promising method to explore overlooked genetic variants in rare diseases with a high suspicion of a genetic basis. We therefore applied PacBio HiFi sequencing in a large multi-generational family presenting with autosomal dominant 46,XY differences of sexual development (DSD), for whom extensive molecular testing over multiple decades had failed to identify a molecular diagnosis. This revealed a rare SINE-VNTR-Alu retroelement insertion in intron 4 of NR5A1, a gene in which loss-of-function variants are an established cause of 46,XY DSD. The insertion segregated among affected family members and was associated with loss-of-expression of alleles in cis, demonstrating a functional impact on NR5A1. This case highlights the power of long-read genome sequencing to detect genomic variants that have previously been intractable to detection by standard short-read genomic testing.


Subject(s)
Disorder of Sex Development, 46,XY , Retroelements , Humans , Mutation , Introns/genetics , Retroelements/genetics , Disorder of Sex Development, 46,XY/genetics , Rare Diseases/genetics , Sexual Development , Steroidogenic Factor 1/genetics
11.
Annu Rev Genomics Hum Genet ; 21: 351-372, 2020 08 31.
Article in English | MEDLINE | ID: mdl-32283948

ABSTRACT

Accurate diagnosis is the cornerstone of medicine; it is essential for informed care and promoting patient and family well-being. However, families with a rare genetic disease (RGD) often spend more than five years on a diagnostic odyssey of specialist visits and invasive testing that is lengthy, costly, and often futile, as 50% of patients do not receive a molecular diagnosis. The current diagnostic paradigm is not well designed for RGDs, especially for patients who remain undiagnosed after the initial set of investigations, and thus requires an expansion of approaches in the clinic. Leveraging opportunities to participate in research programs that utilize new technologies to understand RGDs is an important path forward for patients seeking a diagnosis. Given recent advancements in such technologies and international initiatives, the prospect of identifying a molecular diagnosis for all patients with RGDs has never been so attainable, but achieving this goal will require global cooperation at an unprecedented scale.


Subject(s)
Genome, Human , Genomics/methods , Rare Diseases/diagnosis , Rare Diseases/genetics , Humans
12.
Hum Mutat ; 43(6): 800-811, 2022 06.
Article in English | MEDLINE | ID: mdl-35181971

ABSTRACT

Despite recent progress in the understanding of the genetic etiologies of rare diseases (RDs), a significant number remain intractable to diagnostic and discovery efforts. Broad data collection and sharing of information among RD researchers is therefore critical. In 2018, the Care4Rare Canada Consortium launched the project C4R-SOLVE, a subaim of which was to collect, harmonize, and share both retrospective and prospective Canadian clinical and multiomic data. Here, we introduce Genomics4RD, an integrated web-accessible platform to share Canadian phenotypic and multiomic data between researchers, both within Canada and internationally, for the purpose of discovering the mechanisms that cause RDs. Genomics4RD has been designed to standardize data collection and processing, and to help users systematically collect, prioritize, and visualize participant information. Data storage, authorization, and access procedures have been developed in collaboration with policy experts and stakeholders to ensure the trusted and secure access of data by external researchers. The breadth and standardization of data offered by Genomics4RD allows researchers to compare candidate disease genes and variants between participants (i.e., matchmaking) for discovery purposes, while facilitating the development of computational approaches for multiomic data analyses and enabling clinical translation efforts for new genetic technologies in the future.


Subject(s)
Rare Diseases , Canada , Genetic Association Studies , Humans , Phenotype , Prospective Studies , Rare Diseases/diagnosis , Rare Diseases/genetics , Retrospective Studies
13.
Genet Med ; 24(1): 100-108, 2022 01.
Article in English | MEDLINE | ID: mdl-34906465

ABSTRACT

PURPOSE: Matchmaking has emerged as a useful strategy for building evidence toward causality of novel disease genes in patients with undiagnosed rare diseases. The Matchmaker Exchange (MME) is a collaborative initiative that facilitates international data sharing for matchmaking purposes; however, data on user experience is limited. METHODS: Patients enrolled as part of the Finding of Rare Disease Genes in Canada (FORGE) and Care4Rare Canada research programs had their exome sequencing data reanalyzed by a multidisciplinary research team over a 2-year period. Compelling variants in genes not previously associated with a human phenotype were submitted through the MME node PhenomeCentral, and outcomes were collected. RESULTS: In this study, 194 novel candidate genes were submitted to the MME, resulting in 1514 matches, and 15% of the genes submitted resulted in collaborations. Most submissions resulted in at least 1 match, and most matches were with GeneMatcher (82%), where additional email exchange was required to evaluate the match because of the lack of phenotypic or inheritance information. CONCLUSION: Matchmaking through the MME is an effective way to investigate novel candidate genes; however, it is a labor-intensive process. Engagement from the community to contribute phenotypic, genotypic, and inheritance data will ensure that matchmaking continues to be a useful approach in the future.


Subject(s)
Databases, Genetic , Information Dissemination , Rare Diseases , Canada , Genetic Association Studies , Humans , Information Dissemination/methods , Phenotype , Rare Diseases/diagnosis , Rare Diseases/genetics
14.
Am J Hum Genet ; 103(4): 474-483, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30220433

ABSTRACT

Advances in high-throughput DNA sequencing have revolutionized the discovery of variants in the human genome; however, interpreting the phenotypic effects of those variants is still a challenge. While several computational approaches to predict variant impact are available, their accuracy is limited and further improvement is needed. Here, we introduce ClinPred, an efficient tool for identifying disease-relevant nonsynonymous variants. Our predictor incorporates two machine learning algorithms that use existing pathogenicity scores and, notably, benefits from inclusion of normal population allele frequency from the gnomAD database as an input feature. Another major strength of our approach is the use of ClinVar-a rapidly growing database that allows selection of confidently annotated disease-causing variants-as a training set. Compared to other methods, ClinPred showed superior accuracy for predicting pathogenicity, achieving the highest area under the curve (AUC) score and increasing both the specificity and sensitivity in different test datasets. It also obtained the best performance according to various other metrics. Moreover, ClinPred performance remained robust with respect to disease type (cancer or rare disease) and mechanism (gain or loss of function). Importantly, we observed that adding allele frequency as a predictive feature-as opposed to setting fixed allele frequency cutoffs-boosts the performance of prediction. We provide pre-computed ClinPred scores for all possible human missense variants in the exome to facilitate its use by the community.


Subject(s)
Computational Biology/methods , Disease/genetics , Polymorphism, Single Nucleotide/genetics , Algorithms , Area Under Curve , Exome/genetics , Gene Frequency/genetics , Genome, Human/genetics , High-Throughput Nucleotide Sequencing/methods , Humans , Machine Learning , Software
15.
Am J Hum Genet ; 103(1): 144-153, 2018 07 05.
Article in English | MEDLINE | ID: mdl-29961568

ABSTRACT

Next-generation sequencing has been invaluable in the elucidation of the genetic etiology of many subtypes of intellectual disability in recent years. Here, using exome sequencing and whole-genome sequencing, we identified three de novo truncating mutations in WAS protein family member 1 (WASF1) in five unrelated individuals with moderate to profound intellectual disability with autistic features and seizures. WASF1, also known as WAVE1, is part of the WAVE complex and acts as a mediator between Rac-GTPase and actin to induce actin polymerization. The three mutations connected by Matchmaker Exchange were c.1516C>T (p.Arg506Ter), which occurs in three unrelated individuals, c.1558C>T (p.Gln520Ter), and c.1482delinsGCCAGG (p.Ile494MetfsTer23). All three variants are predicted to partially or fully disrupt the C-terminal actin-binding WCA domain. Functional studies using fibroblast cells from two affected individuals with the c.1516C>T mutation showed a truncated WASF1 and a defect in actin remodeling. This study provides evidence that de novo heterozygous mutations in WASF1 cause a rare form of intellectual disability.


Subject(s)
Intellectual Disability/genetics , Mutation/genetics , Seizures/genetics , Wiskott-Aldrich Syndrome Protein Family/genetics , Adult , Female , Heterozygote , Humans , Male , Exome Sequencing/methods , Young Adult
16.
Am J Hum Genet ; 102(1): 156-174, 2018 01 04.
Article in English | MEDLINE | ID: mdl-29304373

ABSTRACT

Pediatric developmental syndromes present with systemic, complex, and often overlapping clinical features that are not infrequently a consequence of Mendelian inheritance of mutations in genes involved in DNA methylation, establishment of histone modifications, and chromatin remodeling (the "epigenetic machinery"). The mechanistic cross-talk between histone modification and DNA methylation suggests that these syndromes might be expected to display specific DNA methylation signatures that are a reflection of those primary errors associated with chromatin dysregulation. Given the interrelated functions of these chromatin regulatory proteins, we sought to identify DNA methylation epi-signatures that could provide syndrome-specific biomarkers to complement standard clinical diagnostics. In the present study, we examined peripheral blood samples from a large cohort of individuals encompassing 14 Mendelian disorders displaying mutations in the genes encoding proteins of the epigenetic machinery. We demonstrated that specific but partially overlapping DNA methylation signatures are associated with many of these conditions. The degree of overlap among these epi-signatures is minimal, further suggesting that, consistent with the initial event, the downstream changes are unique to every syndrome. In addition, by combining these epi-signatures, we have demonstrated that a machine learning tool can be built to concurrently screen for multiple syndromes with high sensitivity and specificity, and we highlight the utility of this tool in solving ambiguous case subjects presenting with variants of unknown significance, along with its ability to generate accurate predictions for subjects presenting with the overlapping clinical and molecular features associated with the disruption of the epigenetic machinery.


Subject(s)
DNA Methylation/genetics , Genome, Human , Mutation/genetics , Neurodevelopmental Disorders/diagnosis , Neurodevelopmental Disorders/genetics , 5' Untranslated Regions/genetics , Case-Control Studies , Child , Child, Preschool , Cohort Studies , Demography , Epigenesis, Genetic , Humans , Models, Genetic , Neurodevelopmental Disorders/blood , Probability , Reproducibility of Results , Young Adult
17.
Am J Hum Genet ; 103(5): 727-739, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30388400

ABSTRACT

Primary defects in motile cilia result in dysfunction of the apparatus responsible for generating fluid flows. Defects in these mechanisms underlie disorders characterized by poor mucus clearance, resulting in susceptibility to chronic recurrent respiratory infections, often associated with infertility; laterality defects occur in about 50% of such individuals. Here we report biallelic variants in LRRC56 (known as oda8 in Chlamydomonas) identified in three unrelated families. The phenotype comprises laterality defects and chronic pulmonary infections. High-speed video microscopy of cultured epithelial cells from an affected individual showed severely dyskinetic cilia but no obvious ultra-structural abnormalities on routine transmission electron microscopy (TEM). Further investigation revealed that LRRC56 interacts with the intraflagellar transport (IFT) protein IFT88. The link with IFT was interrogated in Trypanosoma brucei. In this protist, LRRC56 is recruited to the cilium during axoneme construction, where it co-localizes with IFT trains and is required for the addition of dynein arms to the distal end of the flagellum. In T. brucei carrying LRRC56-null mutations, or a variant resulting in the p.Leu259Pro substitution corresponding to the p.Leu140Pro variant seen in one of the affected families, we observed abnormal ciliary beat patterns and an absence of outer dynein arms restricted to the distal portion of the axoneme. Together, our findings confirm that deleterious variants in LRRC56 result in a human disease and suggest that this protein has a likely role in dynein transport during cilia assembly that is evolutionarily important for cilia motility.


Subject(s)
Biological Transport/genetics , Flagella/genetics , Mucociliary Clearance/genetics , Mutation/genetics , Proteins/genetics , Adult , Alleles , Axoneme/genetics , Cell Line , Chlamydomonas/genetics , Cilia/genetics , Dyneins/genetics , Epithelial Cells/pathology , Female , HEK293 Cells , Humans , Infant , Male , Phenotype , Trypanosoma brucei brucei/genetics
18.
Am J Med Genet A ; 185(11): 3502-3506, 2021 11.
Article in English | MEDLINE | ID: mdl-34405953

ABSTRACT

Biallelic pathogenic variants in RNU4ATAC have been linked to microcephalic osteodysplastic primordial dwarfism type 1 (MOPD1). Although children with MOPD1 have been reported to show profound, life-limiting clinical decompensation at the time of a febrile illness, these episodes including magnetic resonance imaging (MRI) findings have not been well characterized. We present acute MRI brain findings for a 10-year-old girl with homozygous variants in RNU4ATAC (NR_023343.1) n.55G>A, who presented with two episodes of clinical decompensation associated with a febrile illness in early childhood. The pathogenic variants were identified by whole genome sequencing as RNU4ATAC is not captured in most exome products. Her MRI of the brain revealed symmetric, diffusion restriction of the deep gray nuclei that initially pointed to a mitochondrial disease or acute necrotizing encephalopathy. Her phenotype included microcephaly and profound cognitive impairment that can be seen with MOPD1. However, she did not demonstrate clinical or radiographic evidence of a spondyloepimetaphyseal dysplasia or "primordial dwarfism" that is characteristic of this disease. As such, the predominant neurological presentation of this child represents an atypical variant of RNU4ATAC-associated disease and should be a diagnostic consideration for geneticists and neurologists caring for children, particularly in the event of an acute clinical decline.


Subject(s)
Dwarfism/genetics , Encephalitis/genetics , Fetal Growth Retardation/genetics , Microcephaly/genetics , Osteochondrodysplasias/genetics , RNA, Small Nuclear/genetics , Brain/abnormalities , Brain/diagnostic imaging , Brain/pathology , Child , Child, Preschool , Dwarfism/diagnostic imaging , Dwarfism/pathology , Encephalitis/diagnostic imaging , Encephalitis/pathology , Exome/genetics , Female , Fetal Growth Retardation/diagnostic imaging , Fetal Growth Retardation/pathology , Genetic Predisposition to Disease , Humans , Infant , Infant, Newborn , Magnetic Resonance Imaging , Male , Microcephaly/diagnostic imaging , Microcephaly/pathology , Mutation/genetics , Osteochondrodysplasias/diagnostic imaging , Osteochondrodysplasias/pathology , Phenotype , Exome Sequencing , Whole Genome Sequencing
19.
Am J Med Genet A ; 185(10): 3005-3011, 2021 10.
Article in English | MEDLINE | ID: mdl-34145744

ABSTRACT

WNT9B plays a key role in the development of the mammalian urogenital system. It is essential for the induction of mesonephric and metanephric tubules, the regulation of renal tubule morphogenesis, and the regulation of renal progenitor cell expansion and differentiation. To our knowledge, WNT9B has not been associated with renal defects in humans; however, WNT9B-/- mice have renal agenesis/hypoplasia and reproductive tract abnormalities. We report four individuals from two unrelated consanguineous families with bilateral renal agenesis/hypoplasia/dysplasia and homozygous variants in WNT9B. The proband from Family 1 has bilateral renal cystic dysplasia and chronic kidney disease. He has two deceased siblings who presented with bilateral renal hypoplasia/agenesis. The three affected family members were homozygous for a missense variant in WNT9B (NM_003396.2: c.949G>A/p.(Gly317Arg)). The proband from Family 2 has renal hypoplasia/dysplasia, chronic kidney disease, and is homozygous for a nonsense variant in WNT9B (NM_003396.2: c.11dupC/p.(Pro5Alafs*52)). Two of her siblings died in the neonatal period, one confirmed to be in the context of oligohydramnios. The proband's unaffected brother is also homozygous for the nonsense variant in WNT9B, suggesting nonpenetrance. We propose a novel association of WNT9B and renal anomalies in humans. Further study is needed to delineate the contribution of WNT9B to genitourinary anomalies in humans.


Subject(s)
Congenital Abnormalities/genetics , Kidney Diseases/congenital , Kidney/abnormalities , Urogenital Abnormalities/genetics , Wnt Proteins/genetics , Animals , Child , Congenital Abnormalities/pathology , Female , Homozygote , Humans , Infant , Kidney/pathology , Kidney Diseases/genetics , Kidney Diseases/pathology , Kidney Tubules/growth & development , Kidney Tubules/pathology , Male , Mice , Pregnancy , Urinary Tract/growth & development , Urinary Tract/metabolism , Urinary Tract/pathology , Urogenital Abnormalities/diagnosis , Urogenital Abnormalities/pathology
20.
Can J Neurol Sci ; 48(4): 504-511, 2021 07.
Article in English | MEDLINE | ID: mdl-33059774

ABSTRACT

BACKGROUND: Spinal muscular atrophy (SMA) is characterized by the progressive loss of motor neurons causing muscle atrophy and weakness. Nusinersen, the first effective SMA therapy was approved by Health Canada in June 2017 and has been added to the provincial formulary of all but one Canadian province. Access to this effective therapy has triggered the inclusion of SMA in an increasing number of Newborn Screening (NBS) programs. However, the range of disease-modifying SMN2 gene copy numbers encountered in survival motor neuron 1 (SMN1)-null individuals means that neither screen-positive definition nor resulting treatment decisions can be determined by SMN1 genotype alone. We outline an approach to this challenge, one that specifically addresses the case of SMA newborns with four copies of SMN2. OBJECTIVES: To develop a standardized post-referral evaluation pathway for babies with a positive SMA NBS screen result. METHODS: An SMA NBS pilot trial in Ontario using first-tier MassARRAY and second-tier multi-ligand probe amplification (MLPA) was launched in January 2020. Prior to this, Ontario pediatric neuromuscular disease and NBS experts met to review the evidence regarding the diagnosis and treatment of children with SMA as it pertained to NBS. A post-referral evaluation algorithm was developed, outlining timelines for patient retrieval and management. CONCLUSIONS: Ontario's pilot NBS program has created a standardized path to facilitate early diagnosis of SMA and initiation of treatment. The goal is to provide timely access to those SMA infants in need of therapy to optimize motor function and prolong survival.


Subject(s)
Muscular Atrophy, Spinal , Neonatal Screening , Early Diagnosis , Follow-Up Studies , Humans , Infant, Newborn , Ontario
SELECTION OF CITATIONS
SEARCH DETAIL