Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 561
Filter
Add more filters

Publication year range
1.
Cell ; 177(1): 32-37, 2019 03 21.
Article in English | MEDLINE | ID: mdl-30901545

ABSTRACT

The introduction of exome sequencing in the clinic has sparked tremendous optimism for the future of rare disease diagnosis, and there is exciting opportunity to further leverage these advances. To provide diagnostic clarity to all of these patients, however, there is a critical need for the field to develop and implement strategies to understand the mechanisms underlying all rare diseases and translate these to clinical care.


Subject(s)
Exome Sequencing/trends , Rare Diseases/diagnosis , Translational Research, Biomedical/methods , Exome , Genetic Testing , Genome, Human/genetics , High-Throughput Nucleotide Sequencing/trends , Humans , Rare Diseases/genetics , Sequence Analysis, DNA/methods , Exome Sequencing/methods
2.
Cell ; 176(6): 1310-1324.e10, 2019 03 07.
Article in English | MEDLINE | ID: mdl-30827684

ABSTRACT

DNA rearrangements resulting in human genome structural variants (SVs) are caused by diverse mutational mechanisms. We used long- and short-read sequencing technologies to investigate end products of de novo chromosome 17p11.2 rearrangements and query the molecular mechanisms underlying both recurrent and non-recurrent events. Evidence for an increased rate of clustered single-nucleotide variant (SNV) mutation in cis with non-recurrent rearrangements was found. Indel and SNV formation are associated with both copy-number gains and losses of 17p11.2, occur up to ∼1 Mb away from the breakpoint junctions, and favor C > G transversion substitutions; results suggest that single-stranded DNA is formed during the genesis of the SV and provide compelling support for a microhomology-mediated break-induced replication (MMBIR) mechanism for SV formation. Our data show an additional mutational burden of MMBIR consisting of hypermutation confined to the locus and manifesting as SNVs and indels predominantly within genes.


Subject(s)
Chromosomes, Human, Pair 17 , Mutation , Abnormalities, Multiple/genetics , Chromosome Breakpoints , Chromosome Disorders/genetics , Chromosome Duplication/genetics , DNA Copy Number Variations , DNA Repair/genetics , DNA Replication , Gene Rearrangement , Genome, Human , Genomic Structural Variation , Humans , INDEL Mutation , Models, Genetic , Polymorphism, Single Nucleotide , Recombination, Genetic , Sequence Analysis, DNA/methods , Smith-Magenis Syndrome/genetics
3.
Cell ; 173(4): 864-878.e29, 2018 05 03.
Article in English | MEDLINE | ID: mdl-29681454

ABSTRACT

Diversity in the genetic lesions that cause cancer is extreme. In consequence, a pressing challenge is the development of drugs that target patient-specific disease mechanisms. To address this challenge, we employed a chemistry-first discovery paradigm for de novo identification of druggable targets linked to robust patient selection hypotheses. In particular, a 200,000 compound diversity-oriented chemical library was profiled across a heavily annotated test-bed of >100 cellular models representative of the diverse and characteristic somatic lesions for lung cancer. This approach led to the delineation of 171 chemical-genetic associations, shedding light on the targetability of mechanistic vulnerabilities corresponding to a range of oncogenotypes present in patient populations lacking effective therapy. Chemically addressable addictions to ciliogenesis in TTC21B mutants and GLUT8-dependent serine biosynthesis in KRAS/KEAP1 double mutants are prominent examples. These observations indicate a wealth of actionable opportunities within the complex molecular etiology of cancer.


Subject(s)
Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Lung Neoplasms/pathology , Small Molecule Libraries/pharmacology , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Line, Tumor , Cytochrome P450 Family 4/deficiency , Cytochrome P450 Family 4/genetics , Drug Discovery , G1 Phase Cell Cycle Checkpoints/drug effects , Glucocorticoids/pharmacology , Glucose Transport Proteins, Facilitative/antagonists & inhibitors , Glucose Transport Proteins, Facilitative/genetics , Glucose Transport Proteins, Facilitative/metabolism , Humans , Kelch-Like ECH-Associated Protein 1/genetics , Kelch-Like ECH-Associated Protein 1/metabolism , Lung Neoplasms/metabolism , Microtubule-Associated Proteins/genetics , Microtubule-Associated Proteins/metabolism , Mutation , NF-E2-Related Factor 2/antagonists & inhibitors , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , RNA Interference , RNA, Small Interfering/metabolism , Receptor, Notch2/genetics , Receptor, Notch2/metabolism , Receptors, Glucocorticoid/antagonists & inhibitors , Receptors, Glucocorticoid/genetics , Receptors, Glucocorticoid/metabolism , Small Molecule Libraries/chemistry , Small Molecule Libraries/metabolism
4.
Cell ; 168(5): 830-842.e7, 2017 02 23.
Article in English | MEDLINE | ID: mdl-28235197

ABSTRACT

De novo copy number variants (dnCNVs) arising at multiple loci in a personal genome have usually been considered to reflect cancer somatic genomic instabilities. We describe a multiple dnCNV (MdnCNV) phenomenon in which individuals with genomic disorders carry five to ten constitutional dnCNVs. These CNVs originate from independent formation incidences, are predominantly tandem duplications or complex gains, exhibit breakpoint junction features reminiscent of replicative repair, and show increased de novo point mutations flanking the rearrangement junctions. The active CNV mutation shower appears to be restricted to a transient perizygotic period. We propose that a defect in the CNV formation process is responsible for the "CNV-mutator state," and this state is dampened after early embryogenesis. The constitutional MdnCNV phenomenon resembles chromosomal instability in various cancers. Investigations of this phenomenon may provide unique access to understanding genomic disorders, structural variant mutagenesis, human evolution, and cancer biology.


Subject(s)
Chromosome Aberrations , DNA Copy Number Variations , Genetic Diseases, Inborn/embryology , Genetic Diseases, Inborn/genetics , Genomic Instability , Mutation , Chromosome Breakpoints , Chromosome Duplication , DNA Replication , Embryonic Development , Female , Gametogenesis , Humans , Male
5.
Cell ; 157(3): 636-50, 2014 Apr 24.
Article in English | MEDLINE | ID: mdl-24766809

ABSTRACT

CLP1 is a RNA kinase involved in tRNA splicing. Recently, CLP1 kinase-dead mice were shown to display a neuromuscular disorder with loss of motor neurons and muscle paralysis. Human genome analyses now identified a CLP1 homozygous missense mutation (p.R140H) in five unrelated families, leading to a loss of CLP1 interaction with the tRNA splicing endonuclease (TSEN) complex, largely reduced pre-tRNA cleavage activity, and accumulation of linear tRNA introns. The affected individuals develop severe motor-sensory defects, cortical dysgenesis, and microcephaly. Mice carrying kinase-dead CLP1 also displayed microcephaly and reduced cortical brain volume due to the enhanced cell death of neuronal progenitors that is associated with reduced numbers of cortical neurons. Our data elucidate a neurological syndrome defined by CLP1 mutations that impair tRNA splicing. Reduction of a founder mutation to homozygosity illustrates the importance of rare variations in disease and supports the clan genomics hypothesis.


Subject(s)
Central Nervous System Diseases/genetics , Mutation, Missense , Nuclear Proteins/metabolism , Peripheral Nervous System Diseases/genetics , Phosphotransferases/metabolism , RNA, Transfer/metabolism , Transcription Factors/metabolism , Abnormalities, Multiple/genetics , Abnormalities, Multiple/pathology , Animals , Central Nervous System Diseases/pathology , Cerebrum/pathology , Child, Preschool , Endoribonucleases/metabolism , Female , Fibroblasts/metabolism , Humans , Infant , Male , Mice , Mice, Inbred CBA , Microcephaly/genetics , Peripheral Nervous System Diseases/pathology , RNA, Transfer/genetics , RNA-Binding Proteins
6.
Cell ; 159(1): 200-214, 2014 Sep 25.
Article in English | MEDLINE | ID: mdl-25259927

ABSTRACT

Invertebrate model systems are powerful tools for studying human disease owing to their genetic tractability and ease of screening. We conducted a mosaic genetic screen of lethal mutations on the Drosophila X chromosome to identify genes required for the development, function, and maintenance of the nervous system. We identified 165 genes, most of whose function has not been studied in vivo. In parallel, we investigated rare variant alleles in 1,929 human exomes from families with unsolved Mendelian disease. Genes that are essential in flies and have multiple human homologs were found to be likely to be associated with human diseases. Merging the human data sets with the fly genes allowed us to identify disease-associated mutations in six families and to provide insights into microcephaly associated with brain dysgenesis. This bidirectional synergism between fly genetics and human genomics facilitates the functional annotation of evolutionarily conserved genes involved in human health.


Subject(s)
Disease/genetics , Drosophila melanogaster/genetics , Genetic Testing , Inheritance Patterns , RNA Interference , Animals , Disease Models, Animal , Humans , X Chromosome
7.
Hum Mol Genet ; 33(16): 1429-1441, 2024 Aug 06.
Article in English | MEDLINE | ID: mdl-38747556

ABSTRACT

Inflammation biomarkers can provide valuable insight into the role of inflammatory processes in many diseases and conditions. Sequencing based analyses of such biomarkers can also serve as an exemplar of the genetic architecture of quantitative traits. To evaluate the biological insight, which can be provided by a multi-ancestry, whole-genome based association study, we performed a comprehensive analysis of 21 inflammation biomarkers from up to 38 465 individuals with whole-genome sequencing from the Trans-Omics for Precision Medicine (TOPMed) program (with varying sample size by trait, where the minimum sample size was n = 737 for MMP-1). We identified 22 distinct single-variant associations across 6 traits-E-selectin, intercellular adhesion molecule 1, interleukin-6, lipoprotein-associated phospholipase A2 activity and mass, and P-selectin-that remained significant after conditioning on previously identified associations for these inflammatory biomarkers. We further expanded upon known biomarker associations by pairing the single-variant analysis with a rare variant set-based analysis that further identified 19 significant rare variant set-based associations with 5 traits. These signals were distinct from both significant single variant association signals within TOPMed and genetic signals observed in prior studies, demonstrating the complementary value of performing both single and rare variant analyses when analyzing quantitative traits. We also confirm several previously reported signals from semi-quantitative proteomics platforms. Many of these signals demonstrate the extensive allelic heterogeneity and ancestry-differentiated variant-trait associations common for inflammation biomarkers, a characteristic we hypothesize will be increasingly observed with well-powered, large-scale analyses of complex traits.


Subject(s)
Biomarkers , Genome-Wide Association Study , Inflammation , Precision Medicine , Whole Genome Sequencing , Humans , Precision Medicine/methods , Inflammation/genetics , Genome-Wide Association Study/methods , Whole Genome Sequencing/methods , Polymorphism, Single Nucleotide , Quantitative Trait Loci , Genetic Predisposition to Disease , Female , Interleukin-6/genetics
8.
Am J Hum Genet ; 110(10): 1704-1717, 2023 10 05.
Article in English | MEDLINE | ID: mdl-37802043

ABSTRACT

Long non-coding RNAs (lncRNAs) are known to perform important regulatory functions in lipid metabolism. Large-scale whole-genome sequencing (WGS) studies and new statistical methods for variant set tests now provide an opportunity to assess more associations between rare variants in lncRNA genes and complex traits across the genome. In this study, we used high-coverage WGS from 66,329 participants of diverse ancestries with measurement of blood lipids and lipoproteins (LDL-C, HDL-C, TC, and TG) in the National Heart, Lung, and Blood Institute (NHLBI) Trans-Omics for Precision Medicine (TOPMed) program to investigate the role of lncRNAs in lipid variability. We aggregated rare variants for 165,375 lncRNA genes based on their genomic locations and conducted rare-variant aggregate association tests using the STAAR (variant-set test for association using annotation information) framework. We performed STAAR conditional analysis adjusting for common variants in known lipid GWAS loci and rare-coding variants in nearby protein-coding genes. Our analyses revealed 83 rare lncRNA variant sets significantly associated with blood lipid levels, all of which were located in known lipid GWAS loci (in a ±500-kb window of a Global Lipids Genetics Consortium index variant). Notably, 61 out of 83 signals (73%) were conditionally independent of common regulatory variation and rare protein-coding variation at the same loci. We replicated 34 out of 61 (56%) conditionally independent associations using the independent UK Biobank WGS data. Our results expand the genetic architecture of blood lipids to rare variants in lncRNAs.


Subject(s)
RNA, Long Noncoding , Humans , RNA, Long Noncoding/genetics , Genome-Wide Association Study , Precision Medicine , Whole Genome Sequencing/methods , Lipids/genetics , Polymorphism, Single Nucleotide/genetics
9.
J Immunol ; 212(6): 962-973, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38315012

ABSTRACT

NK cell deficiency (NKD) occurs when an individual's major clinical immunodeficiency derives from abnormal NK cells and is associated with several genetic etiologies. Three categories of ß-actin-related diseases with over 60 ACTB (ß-actin) variants have previously been identified, none with a distinct NK cell phenotype. An individual with mild developmental delay, macrothrombocytopenia, and susceptibility to infections, molluscum contagiosum virus, and EBV-associated lymphoma had functional NKD for over a decade. A de novo ACTB variant encoding G342D ß-actin was identified and was consistent with the individual's developmental and platelet phenotype. This novel variant also was found to have direct impact in NK cells because its expression in the human NK cell line YTS (YTS-NKD) caused increased cell spreading in lytic immune synapses created on activating surfaces. YTS-NKD cells were able to degranulate and perform cytotoxicity, but they demonstrated defective serial killing because of prolonged conjugation to the killed target cell and thus were effectively unable to terminate lytic synapses. G342D ß-actin results in a novel, to our knowledge, mechanism of functional NKD via increased synaptic spreading and defective lytic synapse termination with resulting impaired serial killing, leading to overall reductions in NK cell cytotoxicity.


Subject(s)
Actins , Immunologic Deficiency Syndromes , Humans , Actins/metabolism , Killer Cells, Natural , Cell Line , Blood Platelets/metabolism , Immunologic Deficiency Syndromes/metabolism
10.
Cell ; 147(1): 32-43, 2011 Sep 30.
Article in English | MEDLINE | ID: mdl-21962505

ABSTRACT

Human diseases are caused by alleles that encompass the full range of variant types, from single-nucleotide changes to copy-number variants, and these variations span a broad frequency spectrum, from the very rare to the common. The picture emerging from analysis of whole-genome sequences, the 1000 Genomes Project pilot studies, and targeted genomic sequencing derived from very large sample sizes reveals an abundance of rare and private variants. One implication of this realization is that recent mutation may have a greater influence on disease susceptibility or protection than is conferred by variations that arose in distant ancestors.


Subject(s)
Genetic Predisposition to Disease , Genetics, Population , Genome, Human , Genetic Variation , Genomics , Humans , Pedigree , Pilot Projects , Precision Medicine
11.
Cell ; 144(5): 703-18, 2011 Mar 04.
Article in English | MEDLINE | ID: mdl-21376233

ABSTRACT

Among breast cancers, triple-negative breast cancer (TNBC) is the most poorly understood and is refractory to current targeted therapies. Using a genetic screen, we identify the PTPN12 tyrosine phosphatase as a tumor suppressor in TNBC. PTPN12 potently suppresses mammary epithelial cell proliferation and transformation. PTPN12 is frequently compromised in human TNBCs, and we identify an upstream tumor-suppressor network that posttranscriptionally controls PTPN12. PTPN12 suppresses transformation by interacting with and inhibiting multiple oncogenic tyrosine kinases, including HER2 and EGFR. The tumorigenic and metastatic potential of PTPN12-deficient TNBC cells is severely impaired upon restoration of PTPN12 function or combined inhibition of PTPN12-regulated tyrosine kinases, suggesting that TNBCs are dependent on the proto-oncogenic tyrosine kinases constrained by PTPN12. Collectively, these data identify PTPN12 as a commonly inactivated tumor suppressor and provide a rationale for combinatorially targeting proto-oncogenic tyrosine kinases in TNBC and other cancers based on their profile of tyrosine-phosphatase activity.


Subject(s)
Breast Neoplasms/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 12/genetics , Protein Tyrosine Phosphatase, Non-Receptor Type 12/metabolism , Tumor Suppressor Proteins/metabolism , Breast Neoplasms/drug therapy , Cell Line, Tumor , Cell Transformation, Neoplastic , ErbB Receptors/metabolism , Female , Gene Expression Regulation, Neoplastic , Humans , MAP Kinase Signaling System , MicroRNAs/metabolism , Mutation , Neoplasm Metastasis , Protein Processing, Post-Translational
12.
Nature ; 586(7831): 741-748, 2020 10.
Article in English | MEDLINE | ID: mdl-33116287

ABSTRACT

The African continent is regarded as the cradle of modern humans and African genomes contain more genetic variation than those from any other continent, yet only a fraction of the genetic diversity among African individuals has been surveyed1. Here we performed whole-genome sequencing analyses of 426 individuals-comprising 50 ethnolinguistic groups, including previously unsampled populations-to explore the breadth of genomic diversity across Africa. We uncovered more than 3 million previously undescribed variants, most of which were found among individuals from newly sampled ethnolinguistic groups, as well as 62 previously unreported loci that are under strong selection, which were predominantly found in genes that are involved in viral immunity, DNA repair and metabolism. We observed complex patterns of ancestral admixture and putative-damaging and novel variation, both within and between populations, alongside evidence that Zambia was a likely intermediate site along the routes of expansion of Bantu-speaking populations. Pathogenic variants in genes that are currently characterized as medically relevant were uncommon-but in other genes, variants denoted as 'likely pathogenic' in the ClinVar database were commonly observed. Collectively, these findings refine our current understanding of continental migration, identify gene flow and the response to human disease as strong drivers of genome-level population variation, and underscore the scientific imperative for a broader characterization of the genomic diversity of African individuals to understand human ancestry and improve health.


Subject(s)
Genetic Variation , Genome, Human/genetics , Genomics , Health , Human Migration , Africa/ethnology , DNA Repair/genetics , Datasets as Topic , Female , Gene Flow , Genetics, Medical , Genetics, Population , Health/history , History, Ancient , Human Migration/history , Humans , Immunity/genetics , Language , Male , Metabolism/genetics , Selection, Genetic , Whole Genome Sequencing
13.
PLoS Genet ; 19(11): e1011005, 2023 Nov.
Article in English | MEDLINE | ID: mdl-37934770

ABSTRACT

BACKGROUND: Kinesin motor proteins transport intracellular cargo, including mRNA, proteins, and organelles. Pathogenic variants in kinesin-related genes have been implicated in neurodevelopmental disorders and skeletal dysplasias. We identified de novo, heterozygous variants in KIF5B, encoding a kinesin-1 subunit, in four individuals with osteogenesis imperfecta. The variants cluster within the highly conserved kinesin motor domain and are predicted to interfere with nucleotide binding, although the mechanistic consequences on cell signaling and function are unknown. METHODS: To understand the in vivo genetic mechanism of KIF5B variants, we modeled the p.Thr87Ile variant that was found in two patients in the C. elegans ortholog, unc-116, at the corresponding position (Thr90Ile) by CRISPR/Cas9 editing and performed functional analysis. Next, we studied the cellular and molecular consequences of the recurrent p.Thr87Ile variant by microscopy, RNA and protein analysis in NIH3T3 cells, primary human fibroblasts and bone biopsy. RESULTS: C. elegans heterozygous for the unc-116 Thr90Ile variant displayed abnormal body length and motility phenotypes that were suppressed by additional copies of the wild type allele, consistent with a dominant negative mechanism. Time-lapse imaging of GFP-tagged mitochondria showed defective mitochondria transport in unc-116 Thr90Ile neurons providing strong evidence for disrupted kinesin motor function. Microscopy studies in human cells showed dilated endoplasmic reticulum, multiple intracellular vacuoles, and abnormal distribution of the Golgi complex, supporting an intracellular trafficking defect. RNA sequencing, proteomic analysis, and bone immunohistochemistry demonstrated down regulation of the mTOR signaling pathway that was partially rescued with leucine supplementation in patient cells. CONCLUSION: We report dominant negative variants in the KIF5B kinesin motor domain in individuals with osteogenesis imperfecta. This study expands the spectrum of kinesin-related disorders and identifies dysregulated signaling targets for KIF5B in skeletal development.


Subject(s)
Kinesins , Osteogenesis Imperfecta , Animals , Humans , Mice , Caenorhabditis elegans/genetics , Caenorhabditis elegans/metabolism , Carrier Proteins/genetics , Down-Regulation , Kinesins/genetics , Kinesins/metabolism , NIH 3T3 Cells , Proteomics , Signal Transduction/genetics , TOR Serine-Threonine Kinases/genetics , TOR Serine-Threonine Kinases/metabolism
14.
Hum Mol Genet ; 32(6): 1048-1060, 2023 03 06.
Article in English | MEDLINE | ID: mdl-36444934

ABSTRACT

Diabetic kidney disease (DKD) is recognized as an important public health challenge. However, its genomic mechanisms are poorly understood. To identify rare variants for DKD, we conducted a whole-exome sequencing (WES) study leveraging large cohorts well-phenotyped for chronic kidney disease and diabetes. Our two-stage WES study included 4372 European and African ancestry participants from the Chronic Renal Insufficiency Cohort and Atherosclerosis Risk in Communities studies (stage 1) and 11 487 multi-ancestry Trans-Omics for Precision Medicine participants (stage 2). Generalized linear mixed models, which accounted for genetic relatedness and adjusted for age, sex and ancestry, were used to test associations between single variants and DKD. Gene-based aggregate rare variant analyses were conducted using an optimized sequence kernel association test implemented within our mixed model framework. We identified four novel exome-wide significant DKD-related loci through initiating diabetes. In single-variant analyses, participants carrying a rare, in-frame insertion in the DIS3L2 gene (rs141560952) exhibited a 193-fold increased odds [95% confidence interval (CI): 33.6, 1105] of DKD compared with noncarriers (P = 3.59 × 10-9). Likewise, each copy of a low-frequency KRT6B splice-site variant (rs425827) conferred a 5.31-fold higher odds (95% CI: 3.06, 9.21) of DKD (P = 2.72 × 10-9). Aggregate gene-based analyses further identified ERAP2 (P = 4.03 × 10-8) and NPEPPS (P = 1.51 × 10-7), which are both expressed in the kidney and implicated in renin-angiotensin-aldosterone system modulated immune response. In the largest WES study of DKD, we identified novel rare variant loci attaining exome-wide significance. These findings provide new insights into the molecular mechanisms underlying DKD.


Subject(s)
Diabetes Mellitus , Diabetic Nephropathies , Renal Insufficiency, Chronic , Humans , Aminopeptidases , Diabetic Nephropathies/genetics , Exome Sequencing , Kidney , Renal Insufficiency, Chronic/genetics
15.
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
16.
Am J Hum Genet ; 109(9): 1713-1723, 2022 09 01.
Article in English | MEDLINE | ID: mdl-35948005

ABSTRACT

The leucine-rich glioma-inactivated (LGI) family consists of four highly conserved paralogous genes, LGI1-4, that are highly expressed in mammalian central and/or peripheral nervous systems. LGI1 antibodies are detected in subjects with autoimmune limbic encephalitis and peripheral nerve hyperexcitability syndromes (PNHSs) such as Isaacs and Morvan syndromes. Pathogenic variations of LGI1 and LGI4 are associated with neurological disorders as disease traits including familial temporal lobe epilepsy and neurogenic arthrogryposis multiplex congenita 1 with myelin defects, respectively. No human disease has been reported associated with either LGI2 or LGI3. We implemented exome sequencing and family-based genomics to identify individuals with deleterious variants in LGI3 and utilized GeneMatcher to connect practitioners and researchers worldwide to investigate the clinical and electrophysiological phenotype in affected subjects. We also generated Lgi3-null mice and performed peripheral nerve dissection and immunohistochemistry to examine the juxtaparanode LGI3 microarchitecture. As a result, we identified 16 individuals from eight unrelated families with loss-of-function (LoF) bi-allelic variants in LGI3. Deep phenotypic characterization showed LGI3 LoF causes a potentially clinically recognizable PNHS trait characterized by global developmental delay, intellectual disability, distal deformities with diminished reflexes, visible facial myokymia, and distinctive electromyographic features suggestive of motor nerve instability. Lgi3-null mice showed reduced and mis-localized Kv1 channel complexes in myelinated peripheral axons. Our data demonstrate bi-allelic LoF variants in LGI3 cause a clinically distinguishable disease trait of PNHS, most likely caused by disturbed Kv1 channel distribution in the absence of LGI3.


Subject(s)
Myokymia , Nerve Tissue Proteins , Animals , Autoantibodies , Axons , Genomics , Humans , Intracellular Signaling Peptides and Proteins/genetics , Mammals/genetics , Mice , Nerve Tissue Proteins/genetics , Phenotype , Reverse Genetics
17.
Hum Mol Genet ; 31(18): 3120-3132, 2022 09 10.
Article in English | MEDLINE | ID: mdl-35552711

ABSTRACT

Plasma levels of fibrinogen, coagulation factors VII and VIII and von Willebrand factor (vWF) are four intermediate phenotypes that are heritable and have been associated with the risk of clinical thrombotic events. To identify rare and low-frequency variants associated with these hemostatic factors, we conducted whole-exome sequencing in 10 860 individuals of European ancestry (EA) and 3529 African Americans (AAs) from the Cohorts for Heart and Aging Research in Genomic Epidemiology Consortium and the National Heart, Lung and Blood Institute's Exome Sequencing Project. Gene-based tests demonstrated significant associations with rare variation (minor allele frequency < 5%) in fibrinogen gamma chain (FGG) (with fibrinogen, P = 9.1 × 10-13), coagulation factor VII (F7) (with factor VII, P = 1.3 × 10-72; seven novel variants) and VWF (with factor VIII and vWF; P = 3.2 × 10-14; one novel variant). These eight novel rare variant associations were independent of the known common variants at these loci and tended to have much larger effect sizes. In addition, one of the rare novel variants in F7 was significantly associated with an increased risk of venous thromboembolism in AAs (Ile200Ser; rs141219108; P = 4.2 × 10-5). After restricting gene-based analyses to only loss-of-function variants, a novel significant association was detected and replicated between factor VIII levels and a stop-gain mutation exclusive to AAs (rs3211938) in CD36 molecule (CD36). This variant has previously been linked to dyslipidemia but not with the levels of a hemostatic factor. These efforts represent the largest integration of whole-exome sequence data from two national projects to identify genetic variation associated with plasma hemostatic factors.


Subject(s)
Factor VIII , Hemostatics , Factor VII/genetics , Factor VIII/genetics , Fibrinogen/genetics , Humans , Polymorphism, Single Nucleotide/genetics , Exome Sequencing , von Willebrand Factor/analysis , von Willebrand Factor/genetics
18.
Am J Hum Genet ; 108(7): 1239-1250, 2021 07 01.
Article in English | MEDLINE | ID: mdl-34129815

ABSTRACT

Despite release of the GRCh38 human reference genome more than seven years ago, GRCh37 remains more widely used by most research and clinical laboratories. To date, no study has quantified the impact of utilizing different reference assemblies for the identification of variants associated with rare and common diseases from large-scale exome-sequencing data. By calling variants on both the GRCh37 and GRCh38 references, we identified single-nucleotide variants (SNVs) and insertion-deletions (indels) in 1,572 exomes from participants with Mendelian diseases and their family members. We found that a total of 1.5% of SNVs and 2.0% of indels were discordant when different references were used. Notably, 76.6% of the discordant variants were clustered within discrete discordant reference patches (DISCREPs) comprising only 0.9% of loci targeted by exome sequencing. These DISCREPs were enriched for genomic elements including segmental duplications, fix patch sequences, and loci known to contain alternate haplotypes. We identified 206 genes significantly enriched for discordant variants, most of which were in DISCREPs and caused by multi-mapped reads on the reference assembly that lacked the variant call. Among these 206 genes, eight are implicated in known Mendelian diseases and 53 are associated with common phenotypes from genome-wide association studies. In addition, variant interpretations could also be influenced by the reference after lifting-over variant loci to another assembly. Overall, we identified genes and genomic loci affected by reference assembly choice, including genes associated with Mendelian disorders and complex human diseases that require careful evaluation in both research and clinical applications.


Subject(s)
Exome , Genome, Human , Polymorphism, Single Nucleotide , Cohort Studies , Genetic Diseases, Inborn/genetics , Humans , Reference Values
19.
Am J Hum Genet ; 108(9): 1710-1724, 2021 09 02.
Article in English | MEDLINE | ID: mdl-34450031

ABSTRACT

Coatomer complexes function in the sorting and trafficking of proteins between subcellular organelles. Pathogenic variants in coatomer subunits or associated factors have been reported in multi-systemic disorders, i.e., coatopathies, that can affect the skeletal and central nervous systems. We have identified loss-of-function variants in COPB2, a component of the coatomer complex I (COPI), in individuals presenting with osteoporosis, fractures, and developmental delay of variable severity. Electron microscopy of COPB2-deficient subjects' fibroblasts showed dilated endoplasmic reticulum (ER) with granular material, prominent rough ER, and vacuoles, consistent with an intracellular trafficking defect. We studied the effect of COPB2 deficiency on collagen trafficking because of the critical role of collagen secretion in bone biology. COPB2 siRNA-treated fibroblasts showed delayed collagen secretion with retention of type I collagen in the ER and Golgi and altered distribution of Golgi markers. copb2-null zebrafish embryos showed retention of type II collagen, disorganization of the ER and Golgi, and early larval lethality. Copb2+/- mice exhibited low bone mass, and consistent with the findings in human cells and zebrafish, studies in Copb2+/- mouse fibroblasts suggest ER stress and a Golgi defect. Interestingly, ascorbic acid treatment partially rescued the zebrafish developmental phenotype and the cellular phenotype in Copb2+/- mouse fibroblasts. This work identifies a form of coatopathy due to COPB2 haploinsufficiency, explores a potential therapeutic approach for this disorder, and highlights the role of the COPI complex as a regulator of skeletal homeostasis.


Subject(s)
Bone and Bones/metabolism , Coat Protein Complex I/genetics , Coatomer Protein/genetics , Developmental Disabilities/genetics , Intellectual Disability/genetics , Osteoporosis/genetics , Animals , Ascorbic Acid/pharmacology , Bone and Bones/drug effects , Bone and Bones/pathology , Brain/diagnostic imaging , Brain/drug effects , Brain/metabolism , Brain/pathology , Child , Child, Preschool , Coat Protein Complex I/deficiency , Coatomer Protein/chemistry , Coatomer Protein/deficiency , Collagen Type I/genetics , Collagen Type I/metabolism , Developmental Disabilities/diagnostic imaging , Developmental Disabilities/metabolism , Developmental Disabilities/pathology , Embryo, Nonmammalian , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/metabolism , Endoplasmic Reticulum/pathology , Female , Fibroblasts/drug effects , Fibroblasts/metabolism , Fibroblasts/pathology , Gene Expression Regulation, Developmental , Golgi Apparatus , Haploinsufficiency , Humans , Intellectual Disability/diagnostic imaging , Intellectual Disability/metabolism , Intellectual Disability/pathology , Male , Mice , Osteoporosis/drug therapy , Osteoporosis/metabolism , Osteoporosis/pathology , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Severity of Illness Index , Zebrafish
20.
Am J Hum Genet ; 108(10): 1981-2005, 2021 10 07.
Article in English | MEDLINE | ID: mdl-34582790

ABSTRACT

Neurodevelopmental disorders (NDDs) are clinically and genetically heterogenous; many such disorders are secondary to perturbation in brain development and/or function. The prevalence of NDDs is > 3%, resulting in significant sociocultural and economic challenges to society. With recent advances in family-based genomics, rare-variant analyses, and further exploration of the Clan Genomics hypothesis, there has been a logarithmic explosion in neurogenetic "disease-associated genes" molecular etiology and biology of NDDs; however, the majority of NDDs remain molecularly undiagnosed. We applied genome-wide screening technologies, including exome sequencing (ES) and whole-genome sequencing (WGS), to identify the molecular etiology of 234 newly enrolled subjects and 20 previously unsolved Turkish NDD families. In 176 of the 234 studied families (75.2%), a plausible and genetically parsimonious molecular etiology was identified. Out of 176 solved families, deleterious variants were identified in 218 distinct genes, further documenting the enormous genetic heterogeneity and diverse perturbations in human biology underlying NDDs. We propose 86 candidate disease-trait-associated genes for an NDD phenotype. Importantly, on the basis of objective and internally established variant prioritization criteria, we identified 51 families (51/176 = 28.9%) with multilocus pathogenic variation (MPV), mostly driven by runs of homozygosity (ROHs) - reflecting genomic segments/haplotypes that are identical-by-descent. Furthermore, with the use of additional bioinformatic tools and expansion of ES to additional family members, we established a molecular diagnosis in 5 out of 20 families (25%) who remained undiagnosed in our previously studied NDD cohort emanating from Turkey.


Subject(s)
Genomics/methods , Mutation , Neurodevelopmental Disorders/epidemiology , Phenotype , Adolescent , Adult , Child , Child, Preschool , Cohort Studies , Female , Humans , Infant , Infant, Newborn , Male , Middle Aged , Neurodevelopmental Disorders/genetics , Neurodevelopmental Disorders/pathology , Pedigree , Prevalence , Turkey/epidemiology , Exome Sequencing , Young Adult
SELECTION OF CITATIONS
SEARCH DETAIL