Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 85
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Bioinformatics ; 40(8)2024 Aug 02.
Article in English | MEDLINE | ID: mdl-39128017

ABSTRACT

MOTIVATION: Mendelian randomization (MR) is a widely used approach to estimate causal effect of variation in gene expression on complex traits. Among several MR-based algorithms, transcriptome-wide summary statistics-based Mendelian Randomization approach (TWMR) enables the uses of multiple SNPs as instruments and multiple gene expression traits as exposures to facilitate causal inference in observational studies. RESULTS: Here we present a Python-based implementation of TWMR and revTWMR. Our implementation offers GPU computational support for faster computations and robust computation mode resilient to highly correlated gene expressions and genetic variants. AVAILABILITY AND IMPLEMENTATION: pyTWMR is available at github.com/soreshkov/pyTWMR.


Subject(s)
Mendelian Randomization Analysis , Polymorphism, Single Nucleotide , Software , Transcriptome , Mendelian Randomization Analysis/methods , Transcriptome/genetics , Algorithms , Humans
2.
BMC Genomics ; 25(1): 787, 2024 Aug 14.
Article in English | MEDLINE | ID: mdl-39143522

ABSTRACT

BACKGROUND/OBJECTIVES: This study aims to elucidate the genetic causes of congenital hypogonadotropic hypogonadism (CHH), a rare genetic disorder resulting in GnRH deficiency, in six families from Pakistan. METHODS: Eighteen DNA samples from six families underwent genome sequencing followed by standard evaluation for pathogenic single nucleotide variants (SNVs) and small indels. All families were subsequently analyzed for pathogenic copy number variants (CNVs) using CoverageMaster. RESULTS: Novel pathogenic homozygous SNVs in known CHH genes were identified in four families: two families with variants in GNRHR, and two others harboring KISS1R variants. Subsequent investigation of CNVs in the remaining two families identified novel unique large deletions in ANOS1. CONCLUSION: A combined, systematic analysis of single nucleotide and CNVs helps to improve the diagnostic yield for variants in patients with CHH.


Subject(s)
DNA Copy Number Variations , Hypogonadism , Pedigree , Polymorphism, Single Nucleotide , Humans , Hypogonadism/genetics , Pakistan , Male , Female , Receptors, Kisspeptin-1/genetics , Whole Genome Sequencing , Receptors, LHRH/genetics , Adult , Membrane Proteins/genetics , Nerve Tissue Proteins , Extracellular Matrix Proteins
3.
Brief Bioinform ; 23(2)2022 03 10.
Article in English | MEDLINE | ID: mdl-35224620

ABSTRACT

CoverageMaster (CoM) is a copy number variation (CNV) calling algorithm based on depth-of-coverage maps designed to detect CNVs of any size in exome [whole exome sequencing (WES)] and genome [whole genome sequencing (WGS)] data. The core of the algorithm is the compression of sequencing coverage data in a multiscale Wavelet space and the analysis through an iterative Hidden Markov Model. CoM processes WES and WGS data at nucleotide scale resolution and accurately detects and visualizes full size range CNVs, including single or partial exon deletions and duplications. The results obtained with this approach support the possibility for coverage-based CNV callers to replace probe-based methods such as array comparative genomic hybridization and multiplex ligation-dependent probe amplification in the near future.


Subject(s)
DNA Copy Number Variations , Exome , Comparative Genomic Hybridization/methods , High-Throughput Nucleotide Sequencing/methods , Exome Sequencing , Whole Genome Sequencing
4.
Hum Mol Genet ; 31(1): 1-9, 2021 12 17.
Article in English | MEDLINE | ID: mdl-33693784

ABSTRACT

Knobloch syndrome is an autosomal recessive phenotype mainly characterized by retinal detachment and encephalocele caused by biallelic pathogenic variants in the COL18A1 gene. However, there are patients clinically diagnosed as Knobloch syndrome with unknown molecular etiology not linked to COL18A1. We studied an historical pedigree (published in 1998) designated as KNO2 (Knobloch type 2 syndrome with intellectual disability, autistic behavior, retinal degeneration, encephalocele). Whole exome sequencing of the two affected siblings and the normal parents resulted in the identification of a PAK2 non-synonymous substitution p.(Glu435Lys) as a causative variant. The variant was monoallelic and apparently de novo in both siblings indicating a likely germ-line mosaicism in one of the parents; the mosaicism, however, could not be observed after deep sequencing of blood parental DNA. PAK2 encodes a member of a small group of serine/threonine kinases; these P21-activating kinases (PAKs) are essential in signal transduction and cellular regulation (cytoskeletal dynamics, cell motility, death and survival signaling and cell cycle progression). Structural analysis of the PAK2 p.(Glu435Lys) variant that is located in the kinase domain of the protein predicts a possible compromise in the kinase activity. Functional analysis of the p.(Glu435Lys) PAK2 variant in transfected HEK293T cells results in a partial loss of the kinase activity. PAK2 has been previously suggested as an autism-related gene. Our results show that PAK2-induced phenotypic spectrum is broad and not fully understood. We conclude that the KNO2 syndrome in the studied family is dominant and caused by a deleterious variant in the PAK2 gene.


Subject(s)
Retinal Degeneration , Retinal Detachment , Encephalocele/diagnosis , Encephalocele/genetics , Encephalocele/pathology , HEK293 Cells , Humans , Mutation , Retinal Degeneration/genetics , Retinal Degeneration/pathology , Retinal Detachment/congenital , Retinal Detachment/genetics , p21-Activated Kinases/genetics
5.
Am J Hum Genet ; 106(1): 58-70, 2020 01 02.
Article in English | MEDLINE | ID: mdl-31883645

ABSTRACT

Congenital hypogonadotropic hypogonadism (CHH) is a rare genetic disorder characterized by infertility and the absence of puberty. Defects in GnRH neuron migration or altered GnRH secretion and/or action lead to a severe gonadotropin-releasing hormone (GnRH) deficiency. Given the close developmental association of GnRH neurons with the olfactory primary axons, CHH is often associated with anosmia or hyposmia, in which case it is defined as Kallmann syndrome (KS). The genetics of CHH are heterogeneous, and >40 genes are involved either alone or in combination. Several CHH-related genes controlling GnRH ontogeny encode proteins containing fibronectin-3 (FN3) domains, which are important for brain and neural development. Therefore, we hypothesized that defects in other FN3-superfamily genes would underlie CHH. Next-generation sequencing was performed for 240 CHH unrelated probands and filtered for rare, protein-truncating variants (PTVs) in FN3-superfamily genes. Compared to gnomAD controls the CHH cohort was statistically enriched for PTVs in neuron-derived neurotrophic factor (NDNF) (p = 1.40 × 10-6). Three heterozygous PTVs (p.Lys62∗, p.Tyr128Thrfs∗55, and p.Trp469∗, all absent from the gnomAD database) and an additional heterozygous missense mutation (p.Thr201Ser) were found in four KS probands. Notably, NDNF is expressed along the GnRH neuron migratory route in both mouse embryos and human fetuses and enhances GnRH neuron migration. Further, knock down of the zebrafish ortholog of NDNF resulted in altered GnRH migration. Finally, mice lacking Ndnf showed delayed GnRH neuron migration and altered olfactory axonal projections to the olfactory bulb; both results are consistent with a role of NDNF in GnRH neuron development. Altogether, our results highlight NDNF as a gene involved in the GnRH neuron migration implicated in KS.


Subject(s)
Cell Movement , Hypogonadism/congenital , Hypogonadism/genetics , Mutation , Nerve Growth Factors/genetics , Neurons/pathology , Adolescent , Animals , Cohort Studies , Female , Heterozygote , Humans , Hypogonadism/pathology , Male , Mice , Mice, Knockout , Nerve Growth Factors/physiology , Neurons/metabolism , Pedigree , Zebrafish
6.
Am J Hum Genet ; 107(5): 963-976, 2020 11 05.
Article in English | MEDLINE | ID: mdl-33157009

ABSTRACT

NCKAP1/NAP1 regulates neuronal cytoskeletal dynamics and is essential for neuronal differentiation in the developing brain. Deleterious variants in NCKAP1 have been identified in individuals with autism spectrum disorder (ASD) and intellectual disability; however, its clinical significance remains unclear. To determine its significance, we assemble genotype and phenotype data for 21 affected individuals from 20 unrelated families with predicted deleterious variants in NCKAP1. This includes 16 individuals with de novo (n = 8), transmitted (n = 6), or inheritance unknown (n = 2) truncating variants, two individuals with structural variants, and three with potentially disruptive de novo missense variants. We report a de novo and ultra-rare deleterious variant burden of NCKAP1 in individuals with neurodevelopmental disorders which needs further replication. ASD or autistic features, language and motor delay, and variable expression of intellectual or learning disability are common clinical features. Among inherited cases, there is evidence of deleterious variants segregating with neuropsychiatric disorders. Based on available human brain transcriptomic data, we show that NCKAP1 is broadly and highly expressed in both prenatal and postnatal periods and demostrate enriched expression in excitatory neurons and radial glias but depleted expression in inhibitory neurons. Mouse in utero electroporation experiments reveal that Nckap1 loss of function promotes neuronal migration during early cortical development. Combined, these data support a role for disruptive NCKAP1 variants in neurodevelopmental delay/autism, possibly by interfering with neuronal migration early in cortical development.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Autism Spectrum Disorder/genetics , Intellectual Disability/genetics , Learning Disabilities/genetics , Mutation , Adaptor Proteins, Signal Transducing/deficiency , Adolescent , Animals , Autism Spectrum Disorder/diagnosis , Autism Spectrum Disorder/pathology , Cerebral Cortex/metabolism , Cerebral Cortex/pathology , Child , Female , Gene Expression , Genotype , HEK293 Cells , Humans , Intellectual Disability/diagnosis , Intellectual Disability/pathology , Learning Disabilities/diagnosis , Learning Disabilities/pathology , Male , Mice , Mice, Knockout , Neuroglia/metabolism , Neuroglia/pathology , Neurons/metabolism , Neurons/pathology , Pedigree , Phenotype , Pregnancy , Protein Isoforms/antagonists & inhibitors , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Transcriptome , Young Adult
7.
Am J Hum Genet ; 107(2): 311-324, 2020 08 06.
Article in English | MEDLINE | ID: mdl-32738225

ABSTRACT

Aminoacyl-tRNA synthetases (ARSs) are ubiquitous, ancient enzymes that charge amino acids to cognate tRNA molecules, the essential first step of protein translation. Here, we describe 32 individuals from 21 families, presenting with microcephaly, neurodevelopmental delay, seizures, peripheral neuropathy, and ataxia, with de novo heterozygous and bi-allelic mutations in asparaginyl-tRNA synthetase (NARS1). We demonstrate a reduction in NARS1 mRNA expression as well as in NARS1 enzyme levels and activity in both individual fibroblasts and induced neural progenitor cells (iNPCs). Molecular modeling of the recessive c.1633C>T (p.Arg545Cys) variant shows weaker spatial positioning and tRNA selectivity. We conclude that de novo and bi-allelic mutations in NARS1 are a significant cause of neurodevelopmental disease, where the mechanism for de novo variants could be toxic gain-of-function and for recessive variants, partial loss-of-function.


Subject(s)
Aspartate-tRNA Ligase/genetics , Gain of Function Mutation/genetics , Loss of Function Mutation/genetics , Neurodevelopmental Disorders/genetics , RNA, Transfer, Amino Acyl/genetics , Alleles , Amino Acyl-tRNA Synthetases/genetics , Cell Line , Female , Genetic Predisposition to Disease/genetics , Humans , Male , Pedigree , RNA, Transfer/genetics , Stem Cells/physiology
8.
Hum Mol Genet ; 29(7): 1132-1143, 2020 05 08.
Article in English | MEDLINE | ID: mdl-32129449

ABSTRACT

The molecular cause of the majority of rare autosomal recessive disorders remains unknown. Consanguinity due to extensive homozygosity unravels many recessive phenotypes and facilitates the detection of novel gene-disease links. Here, we report two siblings with phenotypic signs, including intellectual disability (ID), developmental delay and microcephaly from a Pakistani consanguineous family in which we have identified homozygosity for p(Tyr103His) in the PSMB1 gene (Genbank NM_002793) that segregated with the disease phenotype. PSMB1 encodes a ß-type proteasome subunit (i.e. ß6). Modeling of the p(Tyr103His) variant indicates that this variant weakens the interactions between PSMB1/ß6 and PSMA5/α5 proteasome subunits and thus destabilizes the 20S proteasome complex. Biochemical experiments in human SHSY5Y cells revealed that the p(Tyr103His) variant affects both the processing of PSMB1/ß6 and its incorporation into proteasome, thus impairing proteasome activity. CRISPR/Cas9 mutagenesis or morpholino knock-down of the single psmb1 zebrafish orthologue resulted in microcephaly, microphthalmia and reduced brain size. Genetic evidence in the family and functional experiments in human cells and zebrafish indicates that PSMB1/ß6 pathogenic variants are the cause of a recessive disease with ID, microcephaly and developmental delay due to abnormal proteasome assembly.


Subject(s)
Dwarfism/genetics , Microcephaly/genetics , Proteasome Endopeptidase Complex/genetics , Alleles , Animals , Child , Consanguinity , Developmental Disabilities/complications , Developmental Disabilities/genetics , Developmental Disabilities/pathology , Dwarfism/complications , Female , Homozygote , Humans , Intellectual Disability/complications , Intellectual Disability/genetics , Intellectual Disability/pathology , Male , Microcephaly/complications , Microcephaly/pathology , Models, Molecular , Pedigree , Phenotype , Zebrafish/genetics
9.
Hum Mol Genet ; 29(4): 618-623, 2020 03 13.
Article in English | MEDLINE | ID: mdl-31903486

ABSTRACT

In a consanguineous Pakistani family with two affected individuals, a homozygous variant Gly399Val in the eighth transmembrane domain of the taurine transporter SLC6A6 was identified resulting in a hypomorph transporting capacity of ~15% compared with normal. Three-dimensional modeling of this variant has indicated that it likely causes displacement of the Tyr138 (TM3) side chain, important for transport of taurine. The affected individuals presented with rapidly progressive childhood retinal degeneration, cardiomyopathy and almost undetectable plasma taurine levels. Oral taurine supplementation of 100 mg/kg/day resulted in maintenance of normal blood taurine levels. Following approval by the ethics committee, a long-term supplementation treatment was introduced. Remarkably, after 24-months, the cardiomyopathy was corrected in both affected siblings, and in the 6-years-old, the retinal degeneration was arrested, and the vision was clinically improved. Similar therapeutic approaches could be employed in Mendelian phenotypes caused by the dysfunction of the hundreds of other molecular transporters.


Subject(s)
Cardiomyopathies/drug therapy , Membrane Glycoproteins/deficiency , Membrane Transport Proteins/deficiency , Retinal Degeneration/drug therapy , Taurine/therapeutic use , Adolescent , Biological Transport , Cardiomyopathies/metabolism , Cardiomyopathies/pathology , Child , Female , Humans , Male , Pedigree , Retinal Degeneration/metabolism , Retinal Degeneration/pathology
10.
Am J Hum Genet ; 104(6): 1073-1087, 2019 06 06.
Article in English | MEDLINE | ID: mdl-31079899

ABSTRACT

Cargo transport along the cytoplasmic microtubular network is essential for neuronal function, and cytoplasmic dynein-1 is an established molecular motor that is critical for neurogenesis and homeostasis. We performed whole-exome sequencing, homozygosity mapping, and chromosomal microarray studies in five individuals from three independent pedigrees and identified likely-pathogenic variants in DYNC1I2 (Dynein Cytoplasmic 1 Intermediate Chain 2), encoding a component of the cytoplasmic dynein 1 complex. In a consanguineous Pakistani family with three affected individuals presenting with microcephaly, severe intellectual disability, simplification of cerebral gyration, corpus callosum hypoplasia, and dysmorphic facial features, we identified a homozygous splice donor site variant (GenBank: NM_001378.2:c.607+1G>A). We report two additional individuals who have similar neurodevelopmental deficits and craniofacial features and harbor deleterious variants; one individual bears a c.740A>G (p.Tyr247Cys) change in trans with a 374 kb deletion encompassing DYNC1I2, and an unrelated individual harbors the compound-heterozygous variants c.868C>T (p.Gln290∗) and c.740A>G (p.Tyr247Cys). Zebrafish larvae subjected to CRISPR-Cas9 gene disruption or transient suppression of dync1i2a displayed significantly altered craniofacial patterning with concomitant reduction in head size. We monitored cell death and cell cycle progression in dync1i2a zebrafish models and observed significantly increased apoptosis, likely due to prolonged mitosis caused by abnormal spindle morphology, and this finding offers initial insights into the cellular basis of microcephaly. Additionally, complementation studies in zebrafish demonstrate that p.Tyr247Cys attenuates gene function, consistent with protein structural analysis. Our genetic and functional data indicate that DYNC1I2 dysfunction probably causes an autosomal-recessive microcephaly syndrome and highlight further the critical roles of the dynein-1 complex in neurodevelopment.


Subject(s)
Craniofacial Abnormalities/etiology , Dyneins/genetics , Intellectual Disability/etiology , Intracranial Arteriovenous Malformations/etiology , Microcephaly/etiology , Mutation , Zebrafish/growth & development , Adult , Alleles , Amino Acid Sequence , Animals , Child, Preschool , Craniofacial Abnormalities/pathology , Dyneins/chemistry , Dyneins/metabolism , Exome , Female , Homozygote , Humans , Infant , Intellectual Disability/pathology , Intracranial Arteriovenous Malformations/pathology , Male , Microcephaly/pathology , Pedigree , Phenotype , Protein Conformation , Sequence Homology , Exome Sequencing , Young Adult , Zebrafish/genetics , Zebrafish/metabolism
11.
Am J Hum Genet ; 105(5): 907-920, 2019 11 07.
Article in English | MEDLINE | ID: mdl-31607425

ABSTRACT

We report two consanguineous families with probands that exhibit intellectual disability, developmental delay, short stature, aphasia, and hypotonia in which homozygous non-synonymous variants were identified in IQSEC1 (GenBank: NM_001134382.3). In a Pakistani family, the IQSEC1 segregating variant is c.1028C>T (p.Thr343Met), while in a Saudi Arabian family the variant is c.962G>A (p.Arg321Gln). IQSEC1-3 encode guanine nucleotide exchange factors for the small GTPase ARF6 and their loss affects a variety of actin-dependent cellular processes, including AMPA receptor trafficking at synapses. The ortholog of IQSECs in the fly is schizo and its loss affects growth cone guidance at the midline in the CNS, also an actin-dependent process. Overexpression of the reference IQSEC1 cDNA in wild-type flies is lethal, but overexpression of the two variant IQSEC1 cDNAs did not affect viability. Loss of schizo caused embryonic lethality that could be rescued to 2nd instar larvae by moderate expression of the human reference cDNA. However, the p.Arg321Gln and p.Thr343Met variants failed to rescue embryonic lethality. These data indicate that the variants behave as loss-of-function mutations. We also show that schizo in photoreceptors is required for phototransduction. Finally, mice with a conditional Iqsec1 deletion in cortical neurons exhibited an increased density of dendritic spines with an immature morphology. The phenotypic similarity of the affecteds and the functional experiments in flies and mice indicate that IQSEC1 variants are the cause of a recessive disease with intellectual disability, developmental delay, and short stature, and that axonal guidance and dendritic projection defects as well as dendritic spine dysgenesis may underlie disease pathogenesis.


Subject(s)
Developmental Disabilities/genetics , Dwarfism/genetics , Guanine Nucleotide Exchange Factors/genetics , Intellectual Disability/genetics , Mutation/genetics , Adult , Alleles , Animals , Child , Dendritic Spines/genetics , Drosophila/genetics , Female , Humans , Male , Mice , Saudi Arabia , Synapses/genetics , Young Adult
12.
Hum Mol Genet ; 28(6): 972-979, 2019 03 15.
Article in English | MEDLINE | ID: mdl-30481285

ABSTRACT

FBXL3 (F-Box and Leucine Rich Repeat Protein 3) encodes a protein that contains an F-box and several tandem leucine-rich repeats (LRR) domains. FBXL3 is part of the SCF (Skp1-Cullin-F box protein) ubiquitin ligase complex that binds and leads to phosphorylation-dependent degradation of the central clock protein cryptochromes (CRY1 and CRY2) by the proteasome and its absence causes circadian phenotypes in mice and behavioral problems. No FBXL3-related phenotypes have been described in humans. By a combination of exome sequencing and homozygosity mapping, we analyzed two consanguineous families with intellectual disability and identified homozygous loss-of-function (LoF) variants in FBXL3. In the first family, from Pakistan, an FBXL3 frameshift variant [NM_012158.2:c.885delT:p.(Leu295Phefs*25)] was the onlysegregating variant in five affected individuals in two family loops (LOD score: 3.12). In the second family, from Lebanon, we identified a nonsense variant [NM_012158.2:c.445C>T:p.(Arg149*)]. In a third patient from Italy, a likely deleterious non-synonymous variant [NM_012158.2:c.1072T>C:p.(Cys358Arg)] was identified in homozygosity. Protein 3D modeling predicted that the Cys358Arg change influences the binding with CRY2 by destabilizing the structure of the FBXL3, suggesting that this variant is also likely to be LoF. The eight affected individuals from the three families presented with a similar phenotype that included intellectual disability, developmental delay, short stature and mild facial dysmorphism, mainly large nose with a bulbous tip. The phenotypic similarity and the segregation analysis suggest that FBXL3 biallelic, LoF variants link this gene with syndromic autosomal recessive developmental delay/intellectual disability.


Subject(s)
Alleles , Developmental Disabilities/genetics , Dwarfism/genetics , F-Box Proteins/genetics , Genetic Variation , Intellectual Disability/genetics , Adult , Consanguinity , DNA Mutational Analysis , Developmental Disabilities/diagnosis , Dwarfism/diagnosis , F-Box Proteins/chemistry , Facies , Female , Homozygote , Humans , Intellectual Disability/diagnosis , Male , Middle Aged , Models, Molecular , Pedigree , Phenotype , Protein Conformation , Structure-Activity Relationship , Young Adult
13.
Am J Hum Genet ; 103(4): 568-578, 2018 10 04.
Article in English | MEDLINE | ID: mdl-30290152

ABSTRACT

Infantile and childhood-onset cataracts form a heterogeneous group of disorders; among the many genetic causes, numerous pathogenic variants in additional genes associated with autosomal-recessive infantile cataracts remain to be discovered. We identified three consanguineous families affected by bilateral infantile cataracts. Using exome sequencing, we found homozygous loss-of-function variants in DNMBP: nonsense variant c.811C>T (p.Arg271∗) in large family F385 (nine affected individuals; LOD score = 5.18 at θ = 0), frameshift deletion c.2947_2948del (p.Asp983∗) in family F372 (two affected individuals), and frameshift variant c.2852_2855del (p.Thr951Metfs∗41) in family F3 (one affected individual). The phenotypes of all affected individuals include infantile-onset cataracts. RNAi-mediated knockdown of the Drosophila ortholog still life (sif), enriched in lens-secreting cells, affects the development of these cells as well as the localization of E-cadherin, alters the distribution of septate junctions in adjacent cone cells, and leads to a ∼50% reduction in electroretinography amplitudes in young flies. DNMBP regulates the shape of tight junctions, which correspond to the septate junctions in invertebrates, as well as the assembly pattern of E-cadherin in human epithelial cells. E-cadherin has an important role in lens vesicle separation and lens epithelial cell survival in humans. We therefore conclude that DNMBP loss-of-function variants cause infantile-onset cataracts in humans.


Subject(s)
Cataract/genetics , Cytoskeletal Proteins/genetics , Genetic Predisposition to Disease/genetics , Genetic Variation/genetics , Loss of Heterozygosity/genetics , Adult , Alleles , Animals , Cadherins/genetics , Child , Drosophila/genetics , Epithelial Cells/pathology , Exome/genetics , Female , Homozygote , Humans , Lod Score , Male , Middle Aged , Pedigree , Phenotype , Tight Junctions/pathology
14.
Genome Res ; 28(1): 1-10, 2018 01.
Article in English | MEDLINE | ID: mdl-29237728

ABSTRACT

The majority of aneuploid fetuses are spontaneously miscarried. Nevertheless, some aneuploid individuals survive despite the strong genetic insult. Here, we investigate if the survival probability of aneuploid fetuses is affected by the genome-wide burden of slightly deleterious variants. We analyzed two cohorts of live-born Down syndrome individuals (388 genotyped samples and 16 fibroblast transcriptomes) and observed a deficit of slightly deleterious variants on Chromosome 21 and decreased transcriptome-wide variation in the expression level of highly constrained genes. We interpret these results as signatures of embryonic selection, and propose a genetic handicap model whereby an individual bearing an extremely severe deleterious variant (such as aneuploidy) could escape embryonic lethality if the genome-wide burden of slightly deleterious variants is sufficiently low. This approach can be used to study the composition and effect of the numerous slightly deleterious variants in humans and model organisms.


Subject(s)
Aneuploidy , Chromosomes, Human, Pair 21/genetics , Down Syndrome , Genotype , Transcriptome , Abortion, Spontaneous , Down Syndrome/embryology , Down Syndrome/genetics , Female , Humans , Pregnancy
15.
Genet Med ; 23(7): 1246-1254, 2021 07.
Article in English | MEDLINE | ID: mdl-33824500

ABSTRACT

PURPOSE: To elucidate the novel molecular cause in families with a new autosomal recessive neurodevelopmental disorder. METHODS: A combination of exome sequencing and gene matching tools was used to identify pathogenic variants in 17 individuals. Quantitative reverse transcription polymerase chain reaction (RT-qPCR) and subcellular localization studies were used to characterize gene expression profile and localization. RESULTS: Biallelic variants in the TMEM222 gene were identified in 17 individuals from nine unrelated families, presenting with intellectual disability and variable other features, such as aggressive behavior, shy character, body tremors, decreased muscle mass in the lower extremities, and mild hypotonia. We found relatively high TMEM222 expression levels in the human brain, especially in the parietal and occipital cortex. Additionally, subcellular localization analysis in human neurons derived from induced pluripotent stem cells (iPSCs) revealed that TMEM222 localizes to early endosomes in the synapses of mature iPSC-derived neurons. CONCLUSION: Our findings support a role for TMEM222 in brain development and function and adds variants in the gene TMEM222 as a novel underlying cause of an autosomal recessive neurodevelopmental disorder.


Subject(s)
Intellectual Disability , Neurodevelopmental Disorders , Humans , Intellectual Disability/genetics , Neurodevelopmental Disorders/genetics , Pedigree , Exome Sequencing
16.
Haematologica ; 106(8): 2233-2241, 2021 08 01.
Article in English | MEDLINE | ID: mdl-32675224

ABSTRACT

Human immunodeficiency virus (HIV) infection is associated with an increased risk of non-Hodgkin lymphoma (NHL). Even in the era of suppressive antiretroviral treatment, HIV-infected individuals remain at higher risk of developing NHL compared to the general population. To identify potential genetic risk loci, we performed case-control genome-wide association studies and a meta-analysis across three cohorts of HIV+ patients of European ancestry, including a total of 278 cases and 1924 matched controls. We observed a significant association with NHL susceptibility in the C-X-C motif chemokine ligand 12 (CXCL12) region on chromosome 10. A fine mapping analysis identified rs7919208 as the most likely causal variant (P = 4.77e-11), with the G>A polymorphism creating a new transcription factor binding site for BATF and JUND. These results suggest a modulatory role of CXCL12 regulation in the increased susceptibility to NHL observed in the HIV-infected population.


Subject(s)
HIV Infections , Lymphoma, AIDS-Related , Lymphoma, Non-Hodgkin , Anti-Retroviral Agents/therapeutic use , Case-Control Studies , Chemokine CXCL12 , Genome-Wide Association Study , HIV Infections/complications , HIV Infections/drug therapy , HIV Infections/genetics , Humans , Lymphoma, AIDS-Related/drug therapy , Lymphoma, Non-Hodgkin/drug therapy , Lymphoma, Non-Hodgkin/genetics , Polymorphism, Genetic
17.
Nature ; 526(7572): 212-7, 2015 Oct 08.
Article in English | MEDLINE | ID: mdl-26416734

ABSTRACT

HIV-1 Nef, a protein important for the development of AIDS, has well-characterized effects on host membrane trafficking and receptor downregulation. By an unidentified mechanism, Nef increases the intrinsic infectivity of HIV-1 virions in a host-cell-dependent manner. Here we identify the host transmembrane protein SERINC5, and to a lesser extent SERINC3, as a potent inhibitor of HIV-1 particle infectivity that is counteracted by Nef. SERINC5 localizes to the plasma membrane, where it is efficiently incorporated into budding HIV-1 virions and impairs subsequent virion penetration of susceptible target cells. Nef redirects SERINC5 to a Rab7-positive endosomal compartment and thereby excludes it from HIV-1 particles. The ability to counteract SERINC5 was conserved in Nef encoded by diverse primate immunodeficiency viruses, as well as in the structurally unrelated glycosylated Gag from murine leukaemia virus. These examples of functional conservation and convergent evolution emphasize the fundamental importance of SERINC5 as a potent anti-retroviral factor.


Subject(s)
HIV-1/physiology , Host-Pathogen Interactions , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/metabolism , Virion/chemistry , Virion/metabolism , nef Gene Products, Human Immunodeficiency Virus/metabolism , Animals , Cell Line , Cell Membrane/metabolism , Cell Membrane/virology , Endosomes/chemistry , Endosomes/metabolism , Evolution, Molecular , Gene Products, gag/metabolism , Gene Products, nef/chemistry , Gene Products, nef/metabolism , HIV-1/chemistry , Host Specificity , Humans , Leukemia Virus, Murine/chemistry , Leukemia Virus, Murine/physiology , Membrane Glycoproteins , Membrane Proteins/analysis , Neoplasm Proteins/metabolism , Primates/virology , Receptors, Cell Surface/metabolism , rab GTP-Binding Proteins/metabolism , rab7 GTP-Binding Proteins
18.
Proc Natl Acad Sci U S A ; 115(51): 13015-13020, 2018 12 18.
Article in English | MEDLINE | ID: mdl-30510006

ABSTRACT

X-chromosome inactivation (XCI) provides a dosage compensation mechanism where, in each female cell, one of the two X chromosomes is randomly silenced. However, some genes on the inactive X chromosome and outside the pseudoautosomal regions escape from XCI and are expressed from both alleles (escapees). We investigated XCI at single-cell resolution combining deep single-cell RNA sequencing with whole-genome sequencing to examine allelic-specific expression in 935 primary fibroblast and 48 lymphoblastoid single cells from five female individuals. In this framework we integrated an original method to identify and exclude doublets of cells. In fibroblast cells, we have identified 55 genes as escapees including five undescribed escapee genes. Moreover, we observed that all genes exhibit a variable propensity to escape XCI in each cell and cell type and that each cell displays a distinct expression profile of the escapee genes. A metric, the Inactivation Score-defined as the mean of the allelic expression profiles of the escapees per cell-enables us to discover a heterogeneous and continuous degree of cellular XCI with extremes represented by "inactive" cells, i.e., cells exclusively expressing the escaping genes from the active X chromosome and "escaping" cells expressing the escapees from both alleles. We found that this effect is associated with cell-cycle phases and, independently, with the XIST expression level, which is higher in the quiescent phase (G0). Single-cell allele-specific expression is a powerful tool to identify novel escapees in different tissues and provide evidence of an unexpected cellular heterogeneity of XCI.


Subject(s)
Chromosomes, Human, X/genetics , Fibroblasts/metabolism , High-Throughput Nucleotide Sequencing/methods , Single-Cell Analysis/methods , X Chromosome Inactivation , Alleles , Cells, Cultured , Female , Fibroblasts/cytology , Humans , Transcriptome
19.
Hum Mol Genet ; 27(15): 2703-2711, 2018 08 01.
Article in English | MEDLINE | ID: mdl-29771303

ABSTRACT

Developmental eye defects often severely reduce vision. Despite extensive efforts, for a substantial fraction of these cases the molecular causes are unknown. Recessive eye disorders are frequent in consanguineous populations and such large families with multiple affected individuals provide an opportunity to identify recessive causative genes. We studied a Pakistani consanguineous family with three affected individuals with congenital vision loss and progressive eye degeneration. The family was analyzed by exome sequencing of one affected individual and genotyping of all family members. We have identified a non-synonymous homozygous variant (NM_001128918.2: c.1708C > G: p.Arg570Gly) in the MARK3 gene as the likely cause of the phenotype. Given that MARK3 is highly conserved in flies (I: 55%; S: 67%) we knocked down the MARK3 homologue, par-1, in the eye during development. This leads to a significant reduction in eye size, a severe loss of photoreceptors and loss of vision based on electroretinogram (ERG) recordings. Expression of the par-1 p.Arg792Gly mutation (equivalent to the MARK3 variant found in patients) in developing fly eyes also induces loss of eye tissue and reduces the ERG signals. The data in flies and human indicate that the MARK3 variant corresponds to a loss of function. We conclude that the identified mutation in MARK3 establishes a new gene-disease link, since it likely causes structural abnormalities during eye development and visual impairment in humans, and that the function of MARK3/par-1 is evolutionarily conserved in eye development.


Subject(s)
Eye Diseases/genetics , Protein Serine-Threonine Kinases/genetics , Vision Disorders/genetics , Animals , Animals, Genetically Modified , Consanguinity , Drosophila Proteins/genetics , Drosophila Proteins/metabolism , Drosophila melanogaster/genetics , Eye Abnormalities/genetics , Female , Genes, Recessive , Glycogen Synthase Kinase 3/genetics , Glycogen Synthase Kinase 3/metabolism , Humans , Male , Mutation, Missense , Pedigree , Vision Disorders/diagnostic imaging , Exome Sequencing
20.
Am J Hum Genet ; 100(3): 444-453, 2017 Mar 02.
Article in English | MEDLINE | ID: mdl-28190458

ABSTRACT

Genomic imprinting results in parental-specific gene expression. Imprinted genes are involved in the etiology of rare syndromes and have been associated with common diseases such as diabetes and cancer. Standard RNA bulk cell sequencing applied to whole-tissue samples has been used to detect imprinted genes in human and mouse models. However, lowly expressed genes cannot be detected by using RNA bulk approaches. Here, we report an original and robust method that combines single-cell RNA-seq and whole-genome sequencing into an optimized statistical framework to analyze genomic imprinting in specific cell types and in different individuals. Using samples from the probands of 2 family trios and 3 unrelated individuals, 1,084 individual primary fibroblasts were RNA sequenced and more than 700,000 informative heterozygous single-nucleotide variations (SNVs) were genotyped. The allele-specific coverage per gene of each SNV in each single cell was used to fit a beta-binomial distribution to model the likelihood of a gene being expressed from one and the same allele. Genes presenting a significant aggregate allelic ratio (between 0.9 and 1) were retained to identify of the allelic parent of origin. Our approach allowed us to validate the imprinting status of all of the known imprinted genes expressed in fibroblasts and the discovery of nine putative imprinted genes, thereby demonstrating the advantages of single-cell over bulk RNA-seq to identify imprinted genes. The proposed single-cell methodology is a powerful tool for establishing a cell type-specific map of genomic imprinting.


Subject(s)
Alleles , Gene Expression , Genomic Imprinting , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Cell Line , Fibroblasts/cytology , Fibroblasts/metabolism , Genetic Association Studies , Genotyping Techniques , Heterozygote , Humans , Models, Theoretical , Polymorphism, Single Nucleotide , Transcriptome
SELECTION OF CITATIONS
SEARCH DETAIL