Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 34
Filter
1.
Cell ; 182(5): 1214-1231.e11, 2020 09 03.
Article in English | MEDLINE | ID: mdl-32888494

ABSTRACT

Blood cells play essential roles in human health, underpinning physiological processes such as immunity, oxygen transport, and clotting, which when perturbed cause a significant global health burden. Here we integrate data from UK Biobank and a large-scale international collaborative effort, including data for 563,085 European ancestry participants, and discover 5,106 new genetic variants independently associated with 29 blood cell phenotypes covering a range of variation impacting hematopoiesis. We holistically characterize the genetic architecture of hematopoiesis, assess the relevance of the omnigenic model to blood cell phenotypes, delineate relevant hematopoietic cell states influenced by regulatory genetic variants and gene networks, identify novel splice-altering variants mediating the associations, and assess the polygenic prediction potential for blood traits and clinical disorders at the interface of complex and Mendelian genetics. These results show the power of large-scale blood cell trait GWAS to interrogate clinically meaningful variants across a wide allelic spectrum of human variation.


Subject(s)
Genetic Predisposition to Disease/genetics , Multifactorial Inheritance/genetics , Female , Gene Regulatory Networks/genetics , Genome-Wide Association Study/methods , Hematopoiesis/genetics , Humans , Male , Phenotype , Polymorphism, Single Nucleotide/genetics
2.
Am J Hum Genet ; 110(8): 1343-1355, 2023 08 03.
Article in English | MEDLINE | ID: mdl-37541188

ABSTRACT

Despite significant progress in unraveling the genetic causes of neurodevelopmental disorders (NDDs), a substantial proportion of individuals with NDDs remain without a genetic diagnosis after microarray and/or exome sequencing. Here, we aimed to assess the power of short-read genome sequencing (GS), complemented with long-read GS, to identify causal variants in participants with NDD from the National Institute for Health and Care Research (NIHR) BioResource project. Short-read GS was conducted on 692 individuals (489 affected and 203 unaffected relatives) from 465 families. Additionally, long-read GS was performed on five affected individuals who had structural variants (SVs) in technically challenging regions, had complex SVs, or required distal variant phasing. Causal variants were identified in 36% of affected individuals (177/489), and a further 23% (112/489) had a variant of uncertain significance after multiple rounds of re-analysis. Among all reported variants, 88% (333/380) were coding nuclear SNVs or insertions and deletions (indels), and the remainder were SVs, non-coding variants, and mitochondrial variants. Furthermore, long-read GS facilitated the resolution of challenging SVs and invalidated variants of difficult interpretation from short-read GS. This study demonstrates the value of short-read GS, complemented with long-read GS, in investigating the genetic causes of NDDs. GS provides a comprehensive and unbiased method of identifying all types of variants throughout the nuclear and mitochondrial genomes in individuals with NDD.


Subject(s)
Genome, Human , Neurodevelopmental Disorders , Humans , Genome, Human/genetics , Chromosome Mapping , Base Sequence , INDEL Mutation , Neurodevelopmental Disorders/genetics
4.
Nature ; 583(7814): 90-95, 2020 07.
Article in English | MEDLINE | ID: mdl-32499645

ABSTRACT

Primary immunodeficiency (PID) is characterized by recurrent and often life-threatening infections, autoimmunity and cancer, and it poses major diagnostic and therapeutic challenges. Although the most severe forms of PID are identified in early childhood, most patients present in adulthood, typically with no apparent family history and a variable clinical phenotype of widespread immune dysregulation: about 25% of patients have autoimmune disease, allergy is prevalent and up to 10% develop lymphoid malignancies1-3. Consequently, in sporadic (or non-familial) PID genetic diagnosis is difficult and the role of genetics is not well defined. Here we address these challenges by performing whole-genome sequencing in a large PID cohort of 1,318 participants. An analysis of the coding regions of the genome in 886 index cases of PID found that disease-causing mutations in known genes that are implicated in monogenic PID occurred in 10.3% of these patients, and a Bayesian approach (BeviMed4) identified multiple new candidate PID-associated genes, including IVNS1ABP. We also examined the noncoding genome, and found deletions in regulatory regions that contribute to disease causation. In addition, we used a genome-wide association study to identify loci that are associated with PID, and found evidence for the colocalization of-and interplay between-novel high-penetrance monogenic variants and common variants (at the PTPN2 and SOCS1 loci). This begins to explain the contribution of common variants to the variable penetrance and phenotypic complexity that are observed in PID. Thus, using a cohort-based whole-genome-sequencing approach in the diagnosis of PID can increase diagnostic yield and further our understanding of the key pathways that influence immune responsiveness in humans.


Subject(s)
Primary Immunodeficiency Diseases/genetics , Whole Genome Sequencing , Actin-Related Protein 2-3 Complex/genetics , Bayes Theorem , Cohort Studies , Female , Genome-Wide Association Study , Humans , Male , Primary Immunodeficiency Diseases/diagnosis , Primary Immunodeficiency Diseases/immunology , Protein Tyrosine Phosphatase, Non-Receptor Type 2/genetics , RNA-Binding Proteins/genetics , Regulatory Sequences, Nucleic Acid/genetics , Suppressor of Cytokine Signaling 1 Protein/genetics , Transcription Factors/genetics
5.
Blood ; 139(14): 2227-2239, 2022 04 07.
Article in English | MEDLINE | ID: mdl-35051265

ABSTRACT

The process of platelet production has so far been understood to be a 2-stage process: megakaryocyte maturation from hematopoietic stem cells followed by proplatelet formation, with each phase regulating the peripheral blood platelet count. Proplatelet formation releases into the bloodstream beads-on-a-string preplatelets, which undergo fission into mature platelets. For the first time, we show that preplatelet maturation is a third, tightly regulated, critical process akin to cytokinesis that regulates platelet count. We show that deficiency in cytokine receptor-like factor 3 (CRLF3) in mice leads to an isolated and sustained 25% to 48% reduction in the platelet count without any effect on other blood cell lineages. We show that Crlf3-/- preplatelets have increased microtubule stability, possibly because of increased microtubule glutamylation via the interaction of CRLF3 with key members of the Hippo pathway. Using a mouse model of JAK2 V617F essential thrombocythemia, we show that a lack of CRLF3 leads to long-term lineage-specific normalization of the platelet count. We thereby postulate that targeting CRLF3 has therapeutic potential for treatment of thrombocythemia.


Subject(s)
Blood Platelets , Thrombocythemia, Essential , Blood Platelets/metabolism , Humans , Megakaryocytes/metabolism , Microtubules , Platelet Count , Receptors, Cytokine , Thrombocythemia, Essential/drug therapy , Thrombopoiesis/genetics
6.
Blood ; 136(17): 1907-1918, 2020 10 22.
Article in English | MEDLINE | ID: mdl-32573726

ABSTRACT

Hereditary hemorrhagic telangiectasia (HHT) is an autosomal dominant vascular dysplasia. Care delivery for HHT patients is impeded by the need for laborious, repeated phenotyping and gaps in knowledge regarding the relationships between causal DNA variants in ENG, ACVRL1, SMAD4 and GDF2, and clinical manifestations. To address this, we analyzed DNA samples from 183 previously uncharacterized, unrelated HHT and suspected HHT cases using the ThromboGenomics high-throughput sequencing platform. We identified 127 rare variants across 168 heterozygous genotypes. Applying modified American College of Medical Genetics and Genomics Guidelines, 106 variants were classified as pathogenic/likely pathogenic and 21 as nonpathogenic (variant of uncertain significance/benign). Unlike the protein products of ACVRL1 and SMAD4, the extracellular ENG amino acids are not strongly conserved. Our inferences of the functional consequences of causal variants in ENG were therefore informed by the crystal structure of endoglin. We then compared the accuracy of predictions of the causal gene blinded to the genetic data using 2 approaches: subjective clinical predictions and statistical predictions based on 8 Human Phenotype Ontology terms. Both approaches had some predictive power, but they were insufficiently accurate to be used clinically, without genetic testing. The distributions of red cell indices differed by causal gene but not sufficiently for clinical use in isolation from genetic data. We conclude that parallel sequencing of the 4 known HHT genes, multidisciplinary team review of variant calls in the context of detailed clinical information, and statistical and structural modeling improve the prognostication and treatment of HHT.


Subject(s)
Genetic Association Studies , Mutation , Telangiectasia, Hereditary Hemorrhagic/genetics , Activin Receptors, Type II/chemistry , Activin Receptors, Type II/genetics , Cohort Studies , DNA Mutational Analysis/methods , Endoglin/chemistry , Endoglin/genetics , Female , Genetic Association Studies/methods , Genetic Predisposition to Disease , Genetic Testing/methods , Genomics/methods , Growth Differentiation Factor 2/chemistry , Growth Differentiation Factor 2/genetics , Heterozygote , High-Throughput Nucleotide Sequencing , Humans , Male , Models, Molecular , Phenotype , Retrospective Studies , Sequence Analysis, DNA/methods , Smad4 Protein/chemistry , Smad4 Protein/genetics , Telangiectasia, Hereditary Hemorrhagic/epidemiology , Telangiectasia, Hereditary Hemorrhagic/pathology
7.
Blood ; 134(23): 2082-2091, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31064749

ABSTRACT

A targeted high-throughput sequencing (HTS) panel test for clinical diagnostics requires careful consideration of the inclusion of appropriate diagnostic-grade genes, the ability to detect multiple types of genomic variation with high levels of analytic sensitivity and reproducibility, and variant interpretation by a multidisciplinary team (MDT) in the context of the clinical phenotype. We have sequenced 2396 index patients using the ThromboGenomics HTS panel test of diagnostic-grade genes known to harbor variants associated with rare bleeding, thrombotic, or platelet disorders (BTPDs). The molecular diagnostic rate was determined by the clinical phenotype, with an overall rate of 49.2% for all thrombotic, coagulation, platelet count, and function disorder patients and a rate of 3.2% for patients with unexplained bleeding disorders characterized by normal hemostasis test results. The MDT classified 745 unique variants, including copy number variants (CNVs) and intronic variants, as pathogenic, likely pathogenic, or variants of uncertain significance. Half of these variants (50.9%) are novel and 41 unique variants were identified in 7 genes recently found to be implicated in BTPDs. Inspection of canonical hemostasis pathways identified 29 patients with evidence of oligogenic inheritance. A molecular diagnosis has been reported for 894 index patients providing evidence that introducing an HTS genetic test is a valuable addition to laboratory diagnostics in patients with a high likelihood of having an inherited BTPD.


Subject(s)
Blood Platelet Disorders , Hemorrhage , High-Throughput Nucleotide Sequencing , Thrombosis , Blood Platelet Disorders/diagnosis , Blood Platelet Disorders/genetics , Female , Gene Dosage , Hemorrhage/diagnosis , Hemorrhage/genetics , Hemostasis/genetics , Humans , Male , Thrombosis/diagnosis , Thrombosis/genetics
8.
Blood ; 134(23): 2070-2081, 2019 12 05.
Article in English | MEDLINE | ID: mdl-31217188

ABSTRACT

To identify novel causes of hereditary thrombocytopenia, we performed a genetic association analysis of whole-genome sequencing data from 13 037 individuals enrolled in the National Institute for Health Research (NIHR) BioResource, including 233 cases with isolated thrombocytopenia. We found an association between rare variants in the transcription factor-encoding gene IKZF5 and thrombocytopenia. We report 5 causal missense variants in or near IKZF5 zinc fingers, of which 2 occurred de novo and 3 co-segregated in 3 pedigrees. A canonical DNA-zinc finger binding model predicts that 3 of the variants alter DNA recognition. Expression studies showed that chromatin binding was disrupted in mutant compared with wild-type IKZF5, and electron microscopy revealed a reduced quantity of α granules in normally sized platelets. Proplatelet formation was reduced in megakaryocytes from 7 cases relative to 6 controls. Comparison of RNA-sequencing data from platelets, monocytes, neutrophils, and CD4+ T cells from 3 cases and 14 healthy controls showed 1194 differentially expressed genes in platelets but only 4 differentially expressed genes in each of the other blood cell types. In conclusion, IKZF5 is a novel transcriptional regulator of megakaryopoiesis and the eighth transcription factor associated with dominant thrombocytopenia in humans.


Subject(s)
Blood Platelets , Genetic Diseases, Inborn , Germ-Line Mutation , Ikaros Transcription Factor , Mutation, Missense , Thrombocytopenia , Thrombopoiesis/genetics , Blood Platelets/metabolism , Blood Platelets/ultrastructure , Chromatin/genetics , Chromatin/metabolism , Chromatin/ultrastructure , Cytoplasmic Granules/genetics , Cytoplasmic Granules/metabolism , Cytoplasmic Granules/ultrastructure , Female , Gene Expression Regulation , Genetic Diseases, Inborn/genetics , Genetic Diseases, Inborn/metabolism , Genetic Diseases, Inborn/pathology , HEK293 Cells , Humans , Ikaros Transcription Factor/genetics , Ikaros Transcription Factor/metabolism , Male , Thrombocytopenia/genetics , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
9.
J Am Soc Nephrol ; 31(2): 365-373, 2020 02.
Article in English | MEDLINE | ID: mdl-31919107

ABSTRACT

BACKGROUND: Primary membranoproliferative GN, including complement 3 (C3) glomerulopathy, is a rare, untreatable kidney disease characterized by glomerular complement deposition. Complement gene mutations can cause familial C3 glomerulopathy, and studies have reported rare variants in complement genes in nonfamilial primary membranoproliferative GN. METHODS: We analyzed whole-genome sequence data from 165 primary membranoproliferative GN cases and 10,250 individuals without the condition (controls) as part of the National Institutes of Health Research BioResource-Rare Diseases Study. We examined copy number, rare, and common variants. RESULTS: Our analysis included 146 primary membranoproliferative GN cases and 6442 controls who were unrelated and of European ancestry. We observed no significant enrichment of rare variants in candidate genes (genes encoding components of the complement alternative pathway and other genes associated with the related disease atypical hemolytic uremic syndrome; 6.8% in cases versus 5.9% in controls) or exome-wide. However, a significant common variant locus was identified at 6p21.32 (rs35406322) (P=3.29×10-8; odds ratio [OR], 1.93; 95% confidence interval [95% CI], 1.53 to 2.44), overlapping the HLA locus. Imputation of HLA types mapped this signal to a haplotype incorporating DQA1*05:01, DQB1*02:01, and DRB1*03:01 (P=1.21×10-8; OR, 2.19; 95% CI, 1.66 to 2.89). This finding was replicated by analysis of HLA serotypes in 338 individuals with membranoproliferative GN and 15,614 individuals with nonimmune renal failure. CONCLUSIONS: We found that HLA type, but not rare complement gene variation, is associated with primary membranoproliferative GN. These findings challenge the paradigm of complement gene mutations typically causing primary membranoproliferative GN and implicate an underlying autoimmune mechanism in most cases.


Subject(s)
Complement C3/immunology , Glomerulonephritis, Membranoproliferative/genetics , Whole Genome Sequencing , Complement C3 Nephritic Factor/analysis , Female , Glomerulonephritis, Membranoproliferative/etiology , HLA-DQ Antigens/genetics , HLA-DR Antigens/genetics , Humans , Male , Serogroup
10.
Blood ; 129(4): 520-524, 2017 01 26.
Article in English | MEDLINE | ID: mdl-28064200

ABSTRACT

The von Willebrand receptor complex, which is composed of the glycoproteins Ibα, Ibß, GPV, and GPIX, plays an essential role in the earliest steps in hemostasis. During the last 4 decades, it has become apparent that loss of function of any 1 of 3 of the genes encoding these glycoproteins (namely, GP1BA, GP1BB, and GP9) leads to autosomal recessive macrothrombocytopenia complicated by bleeding. A small number of variants in GP1BA have been reported to cause a milder and dominant form of macrothrombocytopenia, but only 2 tentative reports exist of such a variant in GP1BB By analyzing data from a collection of more than 1000 genome-sequenced patients with a rare bleeding and/or platelet disorder, we have identified a significant association between rare monoallelic variants in GP1BB and macrothrombocytopenia. To strengthen our findings, we sought further cases in 2 additional collections in the United Kingdom and Japan. Across 18 families exhibiting phenotypes consistent with autosomal dominant inheritance of macrothrombocytopenia, we report on 27 affected cases carrying 1 of 9 rare variants in GP1BB.


Subject(s)
Blood Platelets/metabolism , Hemorrhage/genetics , Mutation , Platelet Glycoprotein GPIb-IX Complex/genetics , Thrombocytopenia/genetics , Alleles , Blood Platelets/pathology , Case-Control Studies , Female , Gene Expression , Genes, Dominant , Genome, Human , Hemorrhage/diagnosis , Hemorrhage/metabolism , Hemorrhage/pathology , High-Throughput Nucleotide Sequencing , Humans , Male , Pedigree , Platelet Count , Thrombocytopenia/diagnosis , Thrombocytopenia/metabolism , Thrombocytopenia/pathology
11.
J Allergy Clin Immunol ; 142(2): 630-646, 2018 08.
Article in English | MEDLINE | ID: mdl-29391254

ABSTRACT

BACKGROUND: Roifman syndrome is a rare inherited disorder characterized by spondyloepiphyseal dysplasia, growth retardation, cognitive delay, hypogammaglobulinemia, and, in some patients, thrombocytopenia. Compound heterozygous variants in the small nuclear RNA gene RNU4ATAC, which is necessary for U12-type intron splicing, were identified recently as driving Roifman syndrome. OBJECTIVE: We studied 3 patients from 2 unrelated kindreds harboring compound heterozygous or homozygous stem II variants in RNU4ATAC to gain insight into the mechanisms behind this disorder. METHODS: We systematically profiled the immunologic and hematologic compartments of the 3 patients with Roifman syndrome and performed RNA sequencing to unravel important splicing defects in both cell lineages. RESULTS: The patients exhibited a dramatic reduction in B-cell numbers, with differentiation halted at the transitional B-cell stage. Despite abundant B-cell activating factor availability, development past this B-cell activating factor-dependent stage was crippled, with disturbed minor splicing of the critical mitogen-activated protein kinase 1 signaling component. In the hematologic compartment patients with Roifman syndrome demonstrated defects in megakaryocyte differentiation, with inadequate generation of proplatelets. Platelets from patients with Roifman syndrome were rounder, with increased tubulin and actin levels, and contained increased α-granule and dense granule markers. Significant minor intron retention in 354 megakaryocyte genes was observed, including DIAPH1 and HPS1, genes known to regulate platelet and dense granule formation, respectively. CONCLUSION: Together, our results provide novel molecular and cellular data toward understanding the immunologic and hematologic features of Roifman syndrome.


Subject(s)
B-Lymphocytes/physiology , Blood Platelets/physiology , Cardiomyopathies/genetics , Immunologic Deficiency Syndromes/genetics , Megakaryocytes/physiology , Mental Retardation, X-Linked/genetics , Mitogen-Activated Protein Kinase 1/genetics , Osteochondrodysplasias/genetics , Precursor Cells, B-Lymphoid/physiology , RNA, Small Nuclear/genetics , Retinal Diseases/genetics , Adolescent , Cell Differentiation , Cell Lineage , Cell Proliferation , Cells, Cultured , Child , Child, Preschool , Humans , Infant , Mitogen-Activated Protein Kinase 1/metabolism , Pedigree , Primary Immunodeficiency Diseases , Protein Splicing/genetics , Signal Transduction/genetics , Exome Sequencing
12.
Blood ; 127(23): 2903-14, 2016 06 09.
Article in English | MEDLINE | ID: mdl-26912466

ABSTRACT

Macrothrombocytopenia (MTP) is a heterogeneous group of disorders characterized by enlarged and reduced numbers of circulating platelets, sometimes resulting in abnormal bleeding. In most MTP, this phenotype arises because of altered regulation of platelet formation from megakaryocytes (MKs). We report the identification of DIAPH1, which encodes the Rho-effector diaphanous-related formin 1 (DIAPH1), as a candidate gene for MTP using exome sequencing, ontological phenotyping, and similarity regression. We describe 2 unrelated pedigrees with MTP and sensorineural hearing loss that segregate with a DIAPH1 R1213* variant predicting partial truncation of the DIAPH1 diaphanous autoregulatory domain. The R1213* variant was linked to reduced proplatelet formation from cultured MKs, cell clustering, and abnormal cortical filamentous actin. Similarly, in platelets, there was increased filamentous actin and stable microtubules, indicating constitutive activation of DIAPH1. Overexpression of DIAPH1 R1213* in cells reproduced the cytoskeletal alterations found in platelets. Our description of a novel disorder of platelet formation and hearing loss extends the repertoire of DIAPH1-related disease and provides new insight into the autoregulation of DIAPH1 activity.


Subject(s)
Adaptor Proteins, Signal Transducing/genetics , Hearing Loss/genetics , Mutation , Thrombocytopenia/genetics , A549 Cells , Adolescent , Adult , Aged , Case-Control Studies , Cells, Cultured , Child , Female , Formins , Genetic Association Studies , Genetic Predisposition to Disease , HEK293 Cells , Hearing Loss/complications , Humans , Male , Middle Aged , Pedigree , Polymorphism, Single Nucleotide , Syndrome , Thrombocytopenia/complications , Young Adult
13.
Nature ; 453(7199): 1239-43, 2008 Jun 26.
Article in English | MEDLINE | ID: mdl-18488015

ABSTRACT

Recent data from several organisms indicate that the transcribed portions of genomes are larger and more complex than expected, and that many functional properties of transcripts are based not on coding sequences but on regulatory sequences in untranslated regions or non-coding RNAs. Alternative start and polyadenylation sites and regulation of intron splicing add additional dimensions to the rich transcriptional output. This transcriptional complexity has been sampled mainly using hybridization-based methods under one or few experimental conditions. Here we applied direct high-throughput sequencing of complementary DNAs (RNA-Seq), supplemented with data from high-density tiling arrays, to globally sample transcripts of the fission yeast Schizosaccharomyces pombe, independently from available gene annotations. We interrogated transcriptomes under multiple conditions, including rapid proliferation, meiotic differentiation and environmental stress, as well as in RNA processing mutants to reveal the dynamic plasticity of the transcriptional landscape as a function of environmental, developmental and genetic factors. High-throughput sequencing proved to be a powerful and quantitative method to sample transcriptomes deeply at maximal resolution. In contrast to hybridization, sequencing showed little, if any, background noise and was sensitive enough to detect widespread transcription in >90% of the genome, including traces of RNAs that were not robustly transcribed or rapidly degraded. The combined sequencing and strand-specific array data provide rich condition-specific information on novel, mostly non-coding transcripts, untranslated regions and gene structures, thus improving the existing genome annotation. Sequence reads spanning exon-exon or exon-intron junctions give unique insight into a surprising variability in splicing efficiency across introns, genes and conditions. Splicing efficiency was largely coordinated with transcript levels, and increased transcription led to increased splicing in test genes. Hundreds of introns showed such regulated splicing during cellular proliferation or differentiation.


Subject(s)
Eukaryotic Cells/metabolism , Gene Expression Profiling , Oligonucleotide Array Sequence Analysis , Schizosaccharomyces/genetics , Alternative Splicing/genetics , Chromatin Immunoprecipitation , Exons/genetics , Gene Expression Regulation, Fungal , Genes, Fungal/genetics , Introns/genetics , RNA Polymerase II/metabolism , RNA, Fungal/analysis , RNA, Fungal/genetics , RNA, Messenger/analysis , RNA, Messenger/genetics , Schizosaccharomyces pombe Proteins/genetics , Sensitivity and Specificity , Transcription, Genetic/genetics
14.
Nature ; 449(7164): 928-32, 2007 Oct 18.
Article in English | MEDLINE | ID: mdl-17898715

ABSTRACT

Modifications on histones control important biological processes through their effects on chromatin structure. Methylation at lysine 4 on histone H3 (H3K4) is found at the 5' end of active genes and contributes to transcriptional activation by recruiting chromatin-remodelling enzymes. An adjacent arginine residue (H3R2) is also known to be asymmetrically dimethylated (H3R2me2a) in mammalian cells, but its location within genes and its function in transcription are unknown. Here we show that H3R2 is also methylated in budding yeast (Saccharomyces cerevisiae), and by using an antibody specific for H3R2me2a in a chromatin immunoprecipitation-on-chip analysis we determine the distribution of this modification on the entire yeast genome. We find that H3R2me2a is enriched throughout all heterochromatic loci and inactive euchromatic genes and is present at the 3' end of moderately transcribed genes. In all cases the pattern of H3R2 methylation is mutually exclusive with the trimethyl form of H3K4 (H3K4me3). We show that methylation at H3R2 abrogates the trimethylation of H3K4 by the Set1 methyltransferase. The specific effect on H3K4me3 results from the occlusion of Spp1, a Set1 methyltransferase subunit necessary for trimethylation. Thus, the inability of Spp1 to recognize H3 methylated at R2 prevents Set1 from trimethylating H3K4. These results provide the first mechanistic insight into the function of arginine methylation on chromatin.


Subject(s)
Arginine/metabolism , Histones/chemistry , Histones/metabolism , Lysine/metabolism , Saccharomyces cerevisiae/metabolism , Chromatin Immunoprecipitation , DNA-Binding Proteins/chemistry , DNA-Binding Proteins/metabolism , Euchromatin/genetics , Euchromatin/metabolism , Gene Expression Regulation, Fungal , Genes, Fungal/genetics , Genome, Fungal/genetics , Heterochromatin/genetics , Heterochromatin/metabolism , Histone Deacetylases/metabolism , Histone-Lysine N-Methyltransferase , Methylation , Protein Subunits/metabolism , Saccharomyces cerevisiae/genetics , Saccharomyces cerevisiae Proteins/chemistry , Saccharomyces cerevisiae Proteins/metabolism , Shelterin Complex , Silent Information Regulator Proteins, Saccharomyces cerevisiae/metabolism , Sirtuin 2 , Sirtuins/metabolism , Telomere-Binding Proteins/metabolism , Transcription Factors/chemistry , Transcription Factors/metabolism
15.
Nat Genet ; 36(8): 809-17, 2004 Aug.
Article in English | MEDLINE | ID: mdl-15195092

ABSTRACT

Cell-cycle control of transcription seems to be universal, but little is known about its global conservation and biological significance. We report on the genome-wide transcriptional program of the Schizosaccharomyces pombe cell cycle, identifying 407 periodically expressed genes of which 136 show high-amplitude changes. These genes cluster in four major waves of expression. The forkhead protein Sep1p regulates mitotic genes in the first cluster, including Ace2p, which activates transcription in the second cluster during the M-G1 transition and cytokinesis. Other genes in the second cluster, which are required for G1-S progression, are regulated by the MBF complex independently of Sep1p and Ace2p. The third cluster coincides with S phase and a fourth cluster contains genes weakly regulated during G2 phase. Despite conserved cell-cycle transcription factors, differences in regulatory circuits between fission and budding yeasts are evident, revealing evolutionary plasticity of transcriptional control. Periodic transcription of most genes is not conserved between the two yeasts, except for a core set of approximately 40 genes that seem to be universally regulated during the eukaryotic cell cycle and may have key roles in cell-cycle progression.


Subject(s)
Cell Cycle/genetics , Gene Expression Regulation, Fungal , Genes, Fungal , Schizosaccharomyces/genetics , Conserved Sequence , Saccharomyces cerevisiae/genetics
16.
Front Genet ; 13: 888025, 2022.
Article in English | MEDLINE | ID: mdl-35571054

ABSTRACT

There is considerable variability in the susceptibility and progression for COVID-19 and it appears to be strongly correlated with age, gender, ethnicity and pre-existing health conditions. However, to our knowledge, cohort studies of COVID-19 in clinically vulnerable groups are lacking. Host genetics has also emerged as a major risk factor for COVID-19, and variation in the ACE2 receptor, which facilitates entry of the SARS-CoV-2 virus into the cell, has become a major focus of attention. Thus, we interrogated an ethnically diverse cohort of National Health Service (NHS) patients in the United Kingdom (United Kingdom) to assess the association between variants in the ACE2 locus and COVID-19 risk. We analysed whole-genome sequencing (WGS) data of 1,837 cases who were tested positive for SARS-CoV-2, and 37,207 controls who were not tested, from the UK's 100,000 Genomes Project (100KGP) for the presence of ACE2 coding variants and extract expression quantitative trait loci (eQTLs). We identified a splice site variant (rs2285666) associated with increased ACE2 expression with an overrepresentation in SARS-CoV-2 positive patients relative to 100KGP controls (p = 0.015), and in hospitalised European patients relative to outpatients in intra-ethnic comparisons (p = 0.029). We also compared the prevalence of 288 eQTLs, of which 23 were enriched in SARS-CoV-2 positive patients. The eQTL rs12006793 had the largest effect size (d = 0.91), which decreases ACE2 expression and is more prevalent in controls, thus potentially reducing the risk of COVID-19. We identified three novel nonsynonymous variants predicted to alter ACE2 function, and showed that three variants (p.K26R, p. H378R, p. Y515N) alter receptor affinity for the viral Spike (S) protein. Variant p. N720D, more prevalent in the European population (p < 0.001), potentially increases viral entry by affecting the ACE2-TMPRSS2 complex. The spectrum of genetic variants in ACE2 may inform risk stratification of COVID-19 patients and could partially explain the differences in disease susceptibility and severity among different ethnic groups.

17.
Trends Genet ; 24(8): 375-8, 2008 Aug.
Article in English | MEDLINE | ID: mdl-18586348

ABSTRACT

We show that genes with rapidly changing expression levels in response to stress contain significantly lower intron densities in yeasts, thale cress and mice. Therefore, we propose that introns can delay regulatory responses and are selected against in genes whose transcripts require rapid adjustment for survival of environmental challenges. These findings could provide an explanation for the apparent extensive intron loss during the evolution of some eukaryotic lineages.


Subject(s)
Gene Expression Regulation , Introns , Animals , Arabidopsis/genetics , Cell Cycle/genetics , Evolution, Molecular , Mice , Models, Genetic , RNA Splicing , Transcription, Genetic , Yeasts/genetics
20.
J Biomol NMR ; 48(2): 85-92, 2010 Oct.
Article in English | MEDLINE | ID: mdl-20680401

ABSTRACT

We present a suite of software for the complete and easy deposition of NMR data to the PDB and BMRB. This suite uses the CCPN framework and introduces a freely downloadable, graphical desktop application called CcpNmr Entry Completion Interface (ECI) for the secure editing of experimental information and associated datasets through the lifetime of an NMR project. CCPN projects can be created within the CcpNmr Analysis software or by importing existing NMR data files using the CcpNmr FormatConverter. After further data entry and checking with the ECI, the project can then be rapidly deposited to the PDBe using AutoDep, or exported as a complete deposition NMR-STAR file. In full CCPN projects created with ECI, it is straightforward to select chemical shift lists, restraint data sets, structural ensembles and all relevant associated experimental collection details, which all are or will become mandatory when depositing to the PDB. Instructions and download information for the ECI are available from the PDBe web site at http://www.ebi.ac.uk/pdbe/nmr/deposition/eci.html .


Subject(s)
Database Management Systems , Databases, Protein , Nuclear Magnetic Resonance, Biomolecular , Proteins/chemistry , Proteins/classification , User-Computer Interface
SELECTION OF CITATIONS
SEARCH DETAIL